401
|
Cantelmo AR, Pircher A, Kalucka J, Carmeliet P. Vessel pruning or healing: endothelial metabolism as a novel target? Expert Opin Ther Targets 2017; 21:239-247. [PMID: 28081641 PMCID: PMC5526136 DOI: 10.1080/14728222.2017.1282465] [Citation(s) in RCA: 41] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
INTRODUCTION Antiangiogenic drugs were originally designed to starve tumors by cutting off their vascular supply. Unfortunately, when these agents are used as monotherapy or in combination with chemotherapy, they provide only modest survival benefits in the order of weeks to months in most cancer patients. Strategies normalizing the disorganized tumor vasculature offer the potential to increase tumor perfusion and oxygenation, and to improve the efficacy of radio-, chemo- and immunotherapy, while reducing metastasis. Areas covered: This review discusses tumor vascular normalization (TVN) as an alternative strategy for anti-angiogenic cancer treatment. We summarize (pre)-clinical strategies that have been developed to normalize tumor vessels as well as their potential to enhance standard therapy. Notably, we describe how targeting endothelial cell metabolism offers new possibilities for antiangiogenic therapy through evoking TVN. Expert opinion: Several drugs targeting VEGF signaling are now clinically used for antiangiogenic cancer treatment. However, excessive blood vessel pruning impedes perfusion and causes tumor hypoxia, known to promote cancer cell dissemination and impair radio-, chemo- and immunotherapy. Normalized vessels lessen tumor hypoxia, impair cancer cell intravasation and enhance anticancer treatment. New data indicate that targeting endothelial cell metabolism is an alternative strategy of antiangiogenic cancer treatment via promotion of TVN.
Collapse
Affiliation(s)
- Anna Rita Cantelmo
- a Laboratory of Angiogenesis and Vascular Metabolism, Department of Oncology , KU Leuven , Leuven , Belgium.,b Laboratory of Angiogenesis and Vascular Metabolism , Vesalius Research Center, Center for Cancer Biology (CCB), VIB , Leuven , Belgium
| | - Andreas Pircher
- a Laboratory of Angiogenesis and Vascular Metabolism, Department of Oncology , KU Leuven , Leuven , Belgium.,b Laboratory of Angiogenesis and Vascular Metabolism , Vesalius Research Center, Center for Cancer Biology (CCB), VIB , Leuven , Belgium
| | - Joanna Kalucka
- a Laboratory of Angiogenesis and Vascular Metabolism, Department of Oncology , KU Leuven , Leuven , Belgium.,b Laboratory of Angiogenesis and Vascular Metabolism , Vesalius Research Center, Center for Cancer Biology (CCB), VIB , Leuven , Belgium
| | - Peter Carmeliet
- a Laboratory of Angiogenesis and Vascular Metabolism, Department of Oncology , KU Leuven , Leuven , Belgium.,b Laboratory of Angiogenesis and Vascular Metabolism , Vesalius Research Center, Center for Cancer Biology (CCB), VIB , Leuven , Belgium
| |
Collapse
|
402
|
Lupo G, Caporarello N, Olivieri M, Cristaldi M, Motta C, Bramanti V, Avola R, Salmeri M, Nicoletti F, Anfuso CD. Anti-angiogenic Therapy in Cancer: Downsides and New Pivots for Precision Medicine. Front Pharmacol 2017; 7:519. [PMID: 28111549 PMCID: PMC5216034 DOI: 10.3389/fphar.2016.00519] [Citation(s) in RCA: 50] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2016] [Accepted: 12/14/2016] [Indexed: 12/12/2022] Open
Abstract
Primary solid tumors originate close to pre-existing tissue vasculature, initially growing along such tissue blood vessels, and this phenomenon is important for the metastatic potential which frequently occurs in highly vascularized tissues. Unfortunately, preclinic and clinic anti-angiogenic approaches have not been very successful, and multiple factors have been found to contribute to toxicity and tumor resistance. Moreover, tumors can highlight intrinsic or acquired resistances, or show adaptation to the VEGF-targeted therapies. Furthermore, different mechanisms of vascularization, activation of alternative signaling pathways, and increased tumor aggressiveness make this context even more complex. On the other hand, it has to be considered that the transitional restoration of normal, not fenestrated, microvessels allows the drug to reach the tumor and act with the maximum efficiency. However, these effects are time-limited and different, depending on the various types of cancer, and clearly define a specific “normalization window.” So, new horizons in the therapeutic approaches consist on the treatment of the tumor with pro- (instead of anti-) angiogenic therapies, which could strengthen a network of well-structured blood vessels that facilitate the transport of the drug.
Collapse
Affiliation(s)
- Gabriella Lupo
- Department of Biomedical and Biotechnological Sciences, School of Medicine, University of Catania Catania, Italy
| | - Nunzia Caporarello
- Department of Biomedical and Biotechnological Sciences, School of Medicine, University of Catania Catania, Italy
| | - Melania Olivieri
- Department of Biomedical and Biotechnological Sciences, School of Medicine, University of Catania Catania, Italy
| | - Martina Cristaldi
- Department of Biomedical and Biotechnological Sciences, School of Medicine, University of Catania Catania, Italy
| | - Carla Motta
- Department of Biomedical and Biotechnological Sciences, School of Medicine, University of Catania Catania, Italy
| | - Vincenzo Bramanti
- Department of Biomedical and Biotechnological Sciences, School of Medicine, University of Catania Catania, Italy
| | - Roberto Avola
- Department of Biomedical and Biotechnological Sciences, School of Medicine, University of Catania Catania, Italy
| | - Mario Salmeri
- Department of Biomedical and Biotechnological Sciences, School of Medicine, University of Catania Catania, Italy
| | - Ferdinando Nicoletti
- Department of Biomedical and Biotechnological Sciences, School of Medicine, University of Catania Catania, Italy
| | - Carmelina D Anfuso
- Department of Biomedical and Biotechnological Sciences, School of Medicine, University of Catania Catania, Italy
| |
Collapse
|
403
|
Fabian KL, Storkus WJ. Immunotherapeutic Targeting of Tumor-Associated Blood Vessels. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2017; 1036:191-211. [PMID: 29275473 DOI: 10.1007/978-3-319-67577-0_13] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
Pathological angiogenesis occurs during tumor progression and leads in the formation of an abnormal vasculature in the tumor microenvironment (TME). The tumor vasculature is disorganized, tortuous and leaky, resulting in high interstitial pressure and hypoxia in the TME, all of which are events that support tumor growth and survival. Given the sustaining role of the tumor vasculature, it has become an increasingly attractive target for the development of anti-cancer therapies. Antibodies, tyrosine kinase inhibitors and cancer vaccines that target pro-angiogenic factors, angiogenesis-associated receptors or tumor blood vessel-associated antigens continue to be developed and tested for therapeutic efficacy. Preferred anti-angiogenic protocols include those that "normalize" the tumor-associated vasculature which reduce hypoxia and improve tumor blood perfusion, resulting in tumor cell apoptosis, decreased immunosuppression, and enhanced effector immune cell infiltration/tumoricidal action within the TME.
Collapse
Affiliation(s)
- Kellsye L Fabian
- Department of Immunology, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA.
| | - Walter J Storkus
- Department of Immunology, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
- Department of Dermatology, University of Pittsburgh Cancer Institute, Pittsburgh, PA, USA
| |
Collapse
|
404
|
Kim J, de Sampaio PC, Lundy DM, Peng Q, Evans KW, Sugimoto H, Gagea M, Kienast Y, Amaral NSD, Rocha RM, Eikesdal HP, Lønning PE, Meric-Bernstam F, LeBleu VS. Heterogeneous perivascular cell coverage affects breast cancer metastasis and response to chemotherapy. JCI Insight 2016; 1:e90733. [PMID: 28018977 PMCID: PMC5161212 DOI: 10.1172/jci.insight.90733] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022] Open
Abstract
Angiogenesis and co-optive vascular remodeling are prerequisites of solid tumor growth. Vascular heterogeneity, notably perivascular composition, may play a critical role in determining the rate of cancer progression. The contribution of vascular pericyte heterogeneity to cancer progression and therapy response is unknown. Here, we show that angiopoietin-2 (Ang2) orchestrates pericyte heterogeneity in breast cancer with an effect on metastatic disease and response to chemotherapy. Using multispectral imaging of human breast tumor specimens, we report that perivascular composition, as defined by the ratio of PDGFRβ- and desmin+ pericytes, provides information about the response to epirubicin but not paclitaxel. Using 17 distinct patient-derived breast cancer xenografts, we demonstrate a cancer cell-derived influence on stromal Ang2 production and a cancer cell-defined control over tumor vasculature and perivascular heterogeneity. The aggressive features of tumors and their distinct response to therapies may thus emerge by the cancer cell-defined engagement of distinct and heterogeneous angiogenic programs.
Collapse
Affiliation(s)
| | | | | | | | - Kurt W Evans
- Department of Investigational Cancer Therapeutics, and
| | | | - Mihai Gagea
- Department of Veterinary Medicine and Surgery, University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Yvonne Kienast
- Discovery Oncology, Roche Pharmaceutical Research and Early Development, (pRED), Roche Innovation Center, Munich, Germany
| | | | - Rafael Malagoli Rocha
- Molecular Gynecology Laboratory, Gynecology Department, Federal University of São Paulo, Brazil
| | - Hans Petter Eikesdal
- Section of Oncology, Department of Clinical Science, University of Bergen, Bergen, Norway.,Department of Oncology, Haukeland University Hospital, Bergen, Norway
| | - Per Eystein Lønning
- Section of Oncology, Department of Clinical Science, University of Bergen, Bergen, Norway.,Department of Oncology, Haukeland University Hospital, Bergen, Norway
| | - Funda Meric-Bernstam
- Department of Investigational Cancer Therapeutics, and.,Department of Breast Surgical Oncology, University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | | |
Collapse
|
405
|
Niyazi M, Harter PN, Hattingen E, Rottler M, von Baumgarten L, Proescholdt M, Belka C, Lauber K, Mittelbronn M. Bevacizumab and radiotherapy for the treatment of glioblastoma: brothers in arms or unholy alliance? Oncotarget 2016; 7:2313-28. [PMID: 26575171 PMCID: PMC4823037 DOI: 10.18632/oncotarget.6320] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2015] [Accepted: 10/13/2015] [Indexed: 01/05/2023] Open
Abstract
Glioblastoma (GBM) represents the most frequent primary brain tumor in adults and carries a dismal prognosis despite aggressive, multimodal treatment regimens involving maximal resection, radiochemotherapy, and maintenance chemotherapy. Histologically, GBMs are characterized by a high degree of VEGF-mediated vascular proliferation. In consequence, new targeted anti-angiogenic therapies, such as the monoclonal anti-VEGF-A antibody bevacizumab, have proven effective in attenuating tumor (neo)angiogenesis and were shown to possess therapeutic activity in several phase II trials. However, the role of bevacizumab in the context of multimodal therapy approaches appears to be rather complex. This review will give insights into current concepts, limitations, and controversies regarding the molecular mechanisms and the clinical benefits of bevacizumab treatment in combination with radio(chemo)therapy - particularly in face of the results of recent phase III trials, which failed to demonstrate convincing improvements in overall survival (OS).
Collapse
Affiliation(s)
- Maximilian Niyazi
- Department of Radiation Oncology, University of Munich, Germany.,German Cancer Consortium (DKTK) and German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Patrick N Harter
- German Cancer Consortium (DKTK) and German Cancer Research Center (DKFZ), Heidelberg, Germany.,Institute of Neurology (Edinger Institute), Goethe University, Frankfurt, Germany
| | - Elke Hattingen
- Department of Neuroradiology, University Hospital Bonn, Bonn, Germany
| | - Maya Rottler
- Department of Radiation Oncology, University of Munich, Germany.,German Cancer Consortium (DKTK) and German Cancer Research Center (DKFZ), Heidelberg, Germany
| | | | - Martin Proescholdt
- Department of Neurosurgery, University Hospital Regensburg, Regensburg, Germany
| | - Claus Belka
- Department of Radiation Oncology, University of Munich, Germany.,German Cancer Consortium (DKTK) and German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Kirsten Lauber
- Department of Radiation Oncology, University of Munich, Germany.,German Cancer Consortium (DKTK) and German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Michel Mittelbronn
- German Cancer Consortium (DKTK) and German Cancer Research Center (DKFZ), Heidelberg, Germany.,Institute of Neurology (Edinger Institute), Goethe University, Frankfurt, Germany
| |
Collapse
|
406
|
Cantelmo AR, Conradi LC, Brajic A, Goveia J, Kalucka J, Pircher A, Chaturvedi P, Hol J, Thienpont B, Teuwen LA, Schoors S, Boeckx B, Vriens J, Kuchnio A, Veys K, Cruys B, Finotto L, Treps L, Stav-Noraas TE, Bifari F, Stapor P, Decimo I, Kampen K, De Bock K, Haraldsen G, Schoonjans L, Rabelink T, Eelen G, Ghesquière B, Rehman J, Lambrechts D, Malik AB, Dewerchin M, Carmeliet P. Inhibition of the Glycolytic Activator PFKFB3 in Endothelium Induces Tumor Vessel Normalization, Impairs Metastasis, and Improves Chemotherapy. Cancer Cell 2016; 30:968-985. [PMID: 27866851 PMCID: PMC5675554 DOI: 10.1016/j.ccell.2016.10.006] [Citation(s) in RCA: 470] [Impact Index Per Article: 52.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/06/2015] [Revised: 06/30/2016] [Accepted: 10/04/2016] [Indexed: 01/01/2023]
Abstract
Abnormal tumor vessels promote metastasis and impair chemotherapy. Hence, tumor vessel normalization (TVN) is emerging as an anti-cancer treatment. Here, we show that tumor endothelial cells (ECs) have a hyper-glycolytic metabolism, shunting intermediates to nucleotide synthesis. EC haplo-deficiency or blockade of the glycolytic activator PFKFB3 did not affect tumor growth, but reduced cancer cell invasion, intravasation, and metastasis by normalizing tumor vessels, which improved vessel maturation and perfusion. Mechanistically, PFKFB3 inhibition tightened the vascular barrier by reducing VE-cadherin endocytosis in ECs, and rendering pericytes more quiescent and adhesive (via upregulation of N-cadherin) through glycolysis reduction; it also lowered the expression of cancer cell adhesion molecules in ECs by decreasing NF-κB signaling. PFKFB3-blockade treatment also improved chemotherapy of primary and metastatic tumors.
Collapse
Affiliation(s)
- Anna Rita Cantelmo
- Laboratory of Angiogenesis and Vascular Metabolism, Department of Oncology, KU Leuven, Leuven 3000, Belgium; Laboratory of Angiogenesis and Vascular Metabolism, Vesalius Research Center, VIB, Leuven 3000, Belgium
| | - Lena-Christin Conradi
- Laboratory of Angiogenesis and Vascular Metabolism, Department of Oncology, KU Leuven, Leuven 3000, Belgium; Laboratory of Angiogenesis and Vascular Metabolism, Vesalius Research Center, VIB, Leuven 3000, Belgium
| | - Aleksandra Brajic
- Laboratory of Angiogenesis and Vascular Metabolism, Department of Oncology, KU Leuven, Leuven 3000, Belgium; Laboratory of Angiogenesis and Vascular Metabolism, Vesalius Research Center, VIB, Leuven 3000, Belgium
| | - Jermaine Goveia
- Laboratory of Angiogenesis and Vascular Metabolism, Department of Oncology, KU Leuven, Leuven 3000, Belgium; Laboratory of Angiogenesis and Vascular Metabolism, Vesalius Research Center, VIB, Leuven 3000, Belgium
| | - Joanna Kalucka
- Laboratory of Angiogenesis and Vascular Metabolism, Department of Oncology, KU Leuven, Leuven 3000, Belgium; Laboratory of Angiogenesis and Vascular Metabolism, Vesalius Research Center, VIB, Leuven 3000, Belgium
| | - Andreas Pircher
- Laboratory of Angiogenesis and Vascular Metabolism, Department of Oncology, KU Leuven, Leuven 3000, Belgium; Laboratory of Angiogenesis and Vascular Metabolism, Vesalius Research Center, VIB, Leuven 3000, Belgium
| | - Pallavi Chaturvedi
- Department of Pharmacology, University of Illinois College of Medicine, Chicago, IL 60612, USA
| | - Johanna Hol
- Department of Pathology, K.G. Jebsen Inflammation Research Center, Oslo University Hospital, University of Oslo, Oslo 0424, Norway
| | - Bernard Thienpont
- Laboratory for Translational Genetics, Vesalius Research Center, VIB, Leuven 3000, Belgium; Laboratory for Translational Genetics, Department of Oncology, KU Leuven, Leuven 3000, Belgium
| | - Laure-Anne Teuwen
- Laboratory of Angiogenesis and Vascular Metabolism, Department of Oncology, KU Leuven, Leuven 3000, Belgium; Laboratory of Angiogenesis and Vascular Metabolism, Vesalius Research Center, VIB, Leuven 3000, Belgium
| | - Sandra Schoors
- Laboratory of Angiogenesis and Vascular Metabolism, Department of Oncology, KU Leuven, Leuven 3000, Belgium; Laboratory of Angiogenesis and Vascular Metabolism, Vesalius Research Center, VIB, Leuven 3000, Belgium
| | - Bram Boeckx
- Laboratory for Translational Genetics, Vesalius Research Center, VIB, Leuven 3000, Belgium; Laboratory for Translational Genetics, Department of Oncology, KU Leuven, Leuven 3000, Belgium
| | - Joris Vriens
- Laboratory of Ion Channel Research and TRP Research Platform Leuven (TRPLe), KU Leuven, Leuven 3000, Belgium
| | - Anna Kuchnio
- Laboratory of Angiogenesis and Vascular Metabolism, Department of Oncology, KU Leuven, Leuven 3000, Belgium; Laboratory of Angiogenesis and Vascular Metabolism, Vesalius Research Center, VIB, Leuven 3000, Belgium
| | - Koen Veys
- Laboratory of Angiogenesis and Vascular Metabolism, Department of Oncology, KU Leuven, Leuven 3000, Belgium; Laboratory of Angiogenesis and Vascular Metabolism, Vesalius Research Center, VIB, Leuven 3000, Belgium
| | - Bert Cruys
- Laboratory of Angiogenesis and Vascular Metabolism, Department of Oncology, KU Leuven, Leuven 3000, Belgium; Laboratory of Angiogenesis and Vascular Metabolism, Vesalius Research Center, VIB, Leuven 3000, Belgium
| | - Lise Finotto
- Laboratory of Angiogenesis and Vascular Metabolism, Department of Oncology, KU Leuven, Leuven 3000, Belgium; Laboratory of Angiogenesis and Vascular Metabolism, Vesalius Research Center, VIB, Leuven 3000, Belgium
| | - Lucas Treps
- Laboratory of Angiogenesis and Vascular Metabolism, Department of Oncology, KU Leuven, Leuven 3000, Belgium; Laboratory of Angiogenesis and Vascular Metabolism, Vesalius Research Center, VIB, Leuven 3000, Belgium
| | - Tor Espen Stav-Noraas
- Department of Pathology, K.G. Jebsen Inflammation Research Center, Oslo University Hospital, University of Oslo, Oslo 0424, Norway
| | - Francesco Bifari
- Laboratory of Angiogenesis and Vascular Metabolism, Department of Oncology, KU Leuven, Leuven 3000, Belgium; Laboratory of Angiogenesis and Vascular Metabolism, Vesalius Research Center, VIB, Leuven 3000, Belgium
| | - Peter Stapor
- Laboratory of Angiogenesis and Vascular Metabolism, Department of Oncology, KU Leuven, Leuven 3000, Belgium; Laboratory of Angiogenesis and Vascular Metabolism, Vesalius Research Center, VIB, Leuven 3000, Belgium
| | - Ilaria Decimo
- Laboratory of Angiogenesis and Vascular Metabolism, Department of Oncology, KU Leuven, Leuven 3000, Belgium; Laboratory of Angiogenesis and Vascular Metabolism, Vesalius Research Center, VIB, Leuven 3000, Belgium
| | - Kim Kampen
- Laboratory of Angiogenesis and Vascular Metabolism, Department of Oncology, KU Leuven, Leuven 3000, Belgium; Laboratory of Angiogenesis and Vascular Metabolism, Vesalius Research Center, VIB, Leuven 3000, Belgium
| | - Katrien De Bock
- Laboratory of Angiogenesis and Vascular Metabolism, Department of Oncology, KU Leuven, Leuven 3000, Belgium; Laboratory of Angiogenesis and Vascular Metabolism, Vesalius Research Center, VIB, Leuven 3000, Belgium
| | - Guttorm Haraldsen
- Department of Pathology, K.G. Jebsen Inflammation Research Center, Oslo University Hospital, University of Oslo, Oslo 0424, Norway
| | - Luc Schoonjans
- Laboratory of Angiogenesis and Vascular Metabolism, Department of Oncology, KU Leuven, Leuven 3000, Belgium; Laboratory of Angiogenesis and Vascular Metabolism, Vesalius Research Center, VIB, Leuven 3000, Belgium
| | - Ton Rabelink
- Department of Nephrology, Einthoven Laboratory for Vascular Medicine, LUMC, Leiden University Medical Center, Leiden 2300 RC, the Netherlands
| | - Guy Eelen
- Laboratory of Angiogenesis and Vascular Metabolism, Department of Oncology, KU Leuven, Leuven 3000, Belgium; Laboratory of Angiogenesis and Vascular Metabolism, Vesalius Research Center, VIB, Leuven 3000, Belgium
| | - Bart Ghesquière
- Metabolomics Core Facility, Department of Oncology, KU Leuven, Leuven 3000, Belgium; Metabolomics Core Facility, Vesalius Research Center, VIB, Leuven 3000, Belgium
| | - Jalees Rehman
- Department of Pharmacology, University of Illinois College of Medicine, Chicago, IL 60612, USA; Section of Cardiology, Department of Medicine, University of Illinois College of Medicine, Chicago, IL 60612, USA
| | - Diether Lambrechts
- Laboratory for Translational Genetics, Vesalius Research Center, VIB, Leuven 3000, Belgium; Laboratory for Translational Genetics, Department of Oncology, KU Leuven, Leuven 3000, Belgium
| | - Asrar B Malik
- Department of Pharmacology, University of Illinois College of Medicine, Chicago, IL 60612, USA
| | - Mieke Dewerchin
- Laboratory of Angiogenesis and Vascular Metabolism, Department of Oncology, KU Leuven, Leuven 3000, Belgium; Laboratory of Angiogenesis and Vascular Metabolism, Vesalius Research Center, VIB, Leuven 3000, Belgium
| | - Peter Carmeliet
- Laboratory of Angiogenesis and Vascular Metabolism, Department of Oncology, KU Leuven, Leuven 3000, Belgium; Laboratory of Angiogenesis and Vascular Metabolism, Vesalius Research Center, VIB, Leuven 3000, Belgium.
| |
Collapse
|
407
|
Tarhini AA, Tawbi H, Storkus WJ. Vaccine therapy + dasatinib for the treatment of patients with stage IIIB-IV melanoma. Melanoma Manag 2016; 3:251-254. [PMID: 30190895 DOI: 10.2217/mmt-2016-0016] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2016] [Accepted: 06/03/2016] [Indexed: 12/15/2022] Open
Affiliation(s)
- Ahmad A Tarhini
- Department of Medicine, University of Pittsburgh School of Medicine, Pittsburgh, PA 15213, USA.,Melanoma Program of the University of Pittsburgh Cancer Institute, Pittsburgh, PA 15213, USA.,Department of Medicine, University of Pittsburgh School of Medicine, Pittsburgh, PA 15213, USA.,Melanoma Program of the University of Pittsburgh Cancer Institute, Pittsburgh, PA 15213, USA
| | - Hussein Tawbi
- Department of Melanoma Medical Oncology, University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA.,Department of Melanoma Medical Oncology, University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Walter J Storkus
- Melanoma Program of the University of Pittsburgh Cancer Institute, Pittsburgh, PA 15213, USA.,Department of Dermatology, University of Pittsburgh School of Medicine, Pittsburgh, PA 15213, USA.,Department of Immunology, University of Pittsburgh School of Medicine, Pittsburgh, PA 15213, USA.,Department of Pathology, University of Pittsburgh School of Medicine, Pittsburgh, PA 15213, USA.,Department of Bioengineering, University of Pittsburgh School of Medicine, Pittsburgh, PA 15213, USA.,Melanoma Program of the University of Pittsburgh Cancer Institute, Pittsburgh, PA 15213, USA.,Department of Dermatology, University of Pittsburgh School of Medicine, Pittsburgh, PA 15213, USA.,Department of Immunology, University of Pittsburgh School of Medicine, Pittsburgh, PA 15213, USA.,Department of Pathology, University of Pittsburgh School of Medicine, Pittsburgh, PA 15213, USA.,Department of Bioengineering, University of Pittsburgh School of Medicine, Pittsburgh, PA 15213, USA
| |
Collapse
|
408
|
Ruiz-Bañobre J, Anido U, Abdulkader I, Antúnez-López J, López-López R, García-González J. Long-term Response to Nivolumab and Acute Renal Failure in a Patient with Metastatic Papillary Renal Cell Carcinoma and a PD-L1 Tumor Expression Increased with Sunitinib Therapy: A Case Report. Front Oncol 2016; 6:250. [PMID: 27921007 PMCID: PMC5118772 DOI: 10.3389/fonc.2016.00250] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2016] [Accepted: 11/09/2016] [Indexed: 12/31/2022] Open
Abstract
INTRODUCTION Papillary renal cell carcinoma (PRCC), which represents around 20% of renal cell carcinomas, is a heterogeneous disease that includes different tumor types with several clinical and molecular phenotypes. Nivolumab, a fully human IgG4 programed cell death protein 1 immune checkpoint inhibitor antibody, has shown not only an overall survival advantage when compared to everolimus but also a relatively good side-effect profile among patients with previously treated advanced or metastatic renal cell carcinoma. CASE REPORT We describe a case of a young man diagnosed with PRCC that achieved a durable response to nivolumab despite a temporary suspension of the treatment due to a renal function side effect. To the best of our knowledge, it is the first renal failure secondary to nivolumab in a metastatic renal cell carcinoma patient. CONCLUDING REMARKS Nivolumab is a promising drug in patients with metastatic PRCC and long-term responses can be achieved. In case of acute renal failure secondary to this treatment, temporary therapy suspension and a low dose of systemic corticosteroids can recover renal function without a negative impact on treatment efficacy.
Collapse
Affiliation(s)
- Juan Ruiz-Bañobre
- Servizo de Oncoloxía Médica & Grupo de Oncoloxía Médica Traslacional, Instituto de Investigación Sanitaria de Santiago de Compostela (IDIS), Complexo Hospitalario Universitario de Santiago de Compostela, A Coruña, Spain
| | - Urbano Anido
- Servizo de Oncoloxía Médica & Grupo de Oncoloxía Médica Traslacional, Instituto de Investigación Sanitaria de Santiago de Compostela (IDIS), Complexo Hospitalario Universitario de Santiago de Compostela, A Coruña, Spain
| | - Ihab Abdulkader
- Servizo de Anatomía Patolóxica, Instituto de Investigación Sanitaria de Santiago de Compostela (IDIS), Complexo Hospitalario Universitario de Santiago de Compostela, A Coruña, Spain
| | - José Antúnez-López
- Servizo de Anatomía Patolóxica, Instituto de Investigación Sanitaria de Santiago de Compostela (IDIS), Complexo Hospitalario Universitario de Santiago de Compostela, A Coruña, Spain
| | - Rafael López-López
- Servizo de Oncoloxía Médica & Grupo de Oncoloxía Médica Traslacional, Instituto de Investigación Sanitaria de Santiago de Compostela (IDIS), Complexo Hospitalario Universitario de Santiago de Compostela, A Coruña, Spain
| | - Jorge García-González
- Servizo de Oncoloxía Médica & Grupo de Oncoloxía Médica Traslacional, Instituto de Investigación Sanitaria de Santiago de Compostela (IDIS), Complexo Hospitalario Universitario de Santiago de Compostela, A Coruña, Spain
| |
Collapse
|
409
|
Development of a mathematical model to estimate intra-tumor oxygen concentrations through multi-parametric imaging. Biomed Eng Online 2016; 15:114. [PMID: 27733170 PMCID: PMC5062945 DOI: 10.1186/s12938-016-0235-5] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2016] [Accepted: 10/04/2016] [Indexed: 01/22/2023] Open
Abstract
Background Tumor hypoxia is involved in every stage of solid tumor development: formation, progression, metastasis, and apoptosis. Two types of hypoxia exist in tumors—chronic hypoxia and acute hypoxia. Recent studies indicate that the regional hypoxia kinetics is closely linked to metastasis and therapeutic responses, but regional hypoxia kinetics is hard to measure. We propose a novel approach to determine the local pO2 by fusing the parameters obtained from in vivo functional imaging through the use of a modified multivariate Krogh model. Methods To test our idea and its potential to translate into an in vivo setting through the use of existing imaging techniques, simulation studies were performed comparing the local partial oxygen pressure (pO2) from the proposed multivariate image fusion model to the referenced pO2 derived by Green’s function, which considers the contribution from every vessel segment of an entire three-dimensional tumor vasculature to profile tumor oxygen with high spatial resolution. Results pO2 derived from our fusion approach were close to the referenced pO2 with regression slope near 1.0 and an r2 higher than 0.8 if the voxel size (or the spatial resolution set by functional imaging modality) was less than 200 μm. The simulation also showed that the metabolic rate, blood perfusion, and hemoglobin concentration were dominant factors in tissue oxygenation. The impact of the measurement error of functional imaging to the pO2 precision and accuracy was simulated. A Gaussian error function with FWHM equal to 20 % of blood perfusion or fractional vascular volume measurement contributed to average 7 % statistical error in pO2. Conclusion The simulation results indicate that the fusion of multiple parametric maps through the biophysically derived mathematical models can monitor the intra-tumor spatial variations of hypoxia in tumors with existing imaging methods, and the potential to further investigate different forms of hypoxia, such as chronic and acute hypoxia, in response to cancer therapies. Electronic supplementary material The online version of this article (doi:10.1186/s12938-016-0235-5) contains supplementary material, which is available to authorized users.
Collapse
|
410
|
Abbasi AZ, Gordijo CR, Amini MA, Maeda A, Rauth AM, DaCosta RS, Wu XY. Hybrid Manganese Dioxide Nanoparticles Potentiate Radiation Therapy by Modulating Tumor Hypoxia. Cancer Res 2016; 76:6643-6656. [PMID: 27758881 DOI: 10.1158/0008-5472.can-15-3475] [Citation(s) in RCA: 76] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2015] [Revised: 07/15/2016] [Accepted: 08/19/2016] [Indexed: 11/16/2022]
Abstract
Hypoxia in the tumor microenvironment (TME) mediates resistance to radiotherapy and contributes to poor prognosis in patients receiving radiotherapy. Here we report the design of clinically suitable formulations of hybrid manganese dioxide (MnO2) nanoparticles (MDNP) using biocompatible materials to reoxygenate the TME by reacting with endogenous H2O2 MDNP containing hydrophilic terpolymer-protein-MnO2 or hydrophobic polymer-lipid-MnO2 provided different oxygen generation rates in the TME relevant to different clinical settings. In highly hypoxic murine or human xenograft breast tumor models, we found that administering either MDNP formulation before radiotherapy modulated tumor hypoxia and increased radiotherapy efficacy, acting to reduce tumor growth, VEGF expression, and vascular density. MDNP treatment also increased apoptosis and DNA double strand breaks, increasing median host survival 3- to 5-fold. Notably, in the murine model, approximately 40% of tumor-bearing mice were tumor-free after a single treatment with MDNPs plus radiotherapy at a 2.5-fold lower dose than required to achieve the same curative treatment without MDNPs. Overall, our findings offer a preclinical proof of concept for the use of MDNP formulations as effective radiotherapy adjuvants. Cancer Res; 76(22); 6643-56. ©2016 AACR.
Collapse
Affiliation(s)
- Azhar Z Abbasi
- Advanced Pharmaceutics and Drug Delivery Laboratory, Leslie Dan Faculty of Pharmacy, University of Toronto, Toronto, Ontario, Canada
| | - Claudia R Gordijo
- Advanced Pharmaceutics and Drug Delivery Laboratory, Leslie Dan Faculty of Pharmacy, University of Toronto, Toronto, Ontario, Canada
| | - Mohammad Ali Amini
- Advanced Pharmaceutics and Drug Delivery Laboratory, Leslie Dan Faculty of Pharmacy, University of Toronto, Toronto, Ontario, Canada
| | - Azusa Maeda
- Departments of Medical Biophysics and Radiation Oncology, University of Toronto, Toronto, Ontario, Canada
| | - Andrew M Rauth
- Departments of Medical Biophysics and Radiation Oncology, University of Toronto, Toronto, Ontario, Canada
| | - Ralph S DaCosta
- Departments of Medical Biophysics and Radiation Oncology, University of Toronto, Toronto, Ontario, Canada.,Princess Margaret Cancer Center, University Health Network, Toronto, Ontario, Canada.,Techna Institute, University Health Network, Toronto Ontario, Canada
| | - Xiao Yu Wu
- Advanced Pharmaceutics and Drug Delivery Laboratory, Leslie Dan Faculty of Pharmacy, University of Toronto, Toronto, Ontario, Canada.
| |
Collapse
|
411
|
Okkenhaug K, Graupera M, Vanhaesebroeck B. Targeting PI3K in Cancer: Impact on Tumor Cells, Their Protective Stroma, Angiogenesis, and Immunotherapy. Cancer Discov 2016; 6:1090-1105. [PMID: 27655435 PMCID: PMC5293166 DOI: 10.1158/2159-8290.cd-16-0716] [Citation(s) in RCA: 197] [Impact Index Per Article: 21.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2016] [Accepted: 08/02/2016] [Indexed: 12/28/2022]
Abstract
The PI3K pathway is hyperactivated in most cancers, yet the capacity of PI3K inhibitors to induce tumor cell death is limited. The efficacy of PI3K inhibition can also derive from interference with the cancer cells' ability to respond to stromal signals, as illustrated by the approved PI3Kδ inhibitor idelalisib in B-cell malignancies. Inhibition of the leukocyte-enriched PI3Kδ or PI3Kγ may unleash antitumor T-cell responses by inhibiting regulatory T cells and immune-suppressive myeloid cells. Moreover, tumor angiogenesis may be targeted by PI3K inhibitors to enhance cancer therapy. Future work should therefore also explore the effects of PI3K inhibitors on the tumor stroma, in addition to their cancer cell-intrinsic impact. SIGNIFICANCE The PI3K pathway extends beyond the direct regulation of cancer cell proliferation and survival. In B-cell malignancies, targeting PI3K purges the tumor cells from their protective microenvironment. Moreover, we propose that PI3K isoform-selective inhibitors may be exploited in the context of cancer immunotherapy and by targeting angiogenesis to improve drug and immune cell delivery. Cancer Discov; 6(10); 1090-105. ©2016 AACR.
Collapse
Affiliation(s)
- Klaus Okkenhaug
- Laboratory of Lymphocyte Signalling and Development, The Babraham Institute, Babraham Research Campus, Cambridge, United Kingdom.
| | - Mariona Graupera
- Institut d'Investigació Biomèdica de Bellvitge (IDIBELL), L'Hospitalet de Llobregat, Barcelona, Spain.
| | | |
Collapse
|
412
|
Wang X, Wei B, Cheng X, Wang J, Tang R. Phenylboronic acid-decorated gelatin nanoparticles for enhanced tumor targeting and penetration. NANOTECHNOLOGY 2016; 27:385101. [PMID: 27514078 DOI: 10.1088/0957-4484/27/38/385101] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/06/2023]
Abstract
Phenylboronic acid-decorated nanoparticles (NPs) were prepared for tumor-targeted drug delivery. 3-carboxyphenylboronic acid (3-CPBA) was modified on the surface of conventional gelatin NPs (designated as NP1) to give tumor-targeting NPs (designated as NP2). The morphology and stability of NP1 and NP2 were then investigated using transmission electron microscopy, scanning electron microscopy, and dynamic light scattering. The results show that both NP1 and NP2 are spherical-like and kinetically stable under various conditions. Doxorubicin hydrochloride (DOX) was used as a model anticancer drug and was loaded into NP1 (NP1-DOX) and NP2 (NP2-DOX). The i n vitro cellular uptake and cytotoxicity of NP1-DOX and NP2-DOX were measured using SH-SY5Y cells, H22 cells, and HepG2 cells. Tumor penetration, accumulation, and antitumor activity were investigated using SH-SY5Y tumor-like spheroids and H22 tumor-bearing mice. All results demonstrated that the conjugation of 3-CPBA can efficiently enhance non-targeted NPs' tumor-homing activity, thus improving their tumor accumulation and antitumor effect.
Collapse
Affiliation(s)
- Xin Wang
- Engineering Research Center for Biomedical Materials, School of Life Science, Anhui University, Hefei 230601, People's Republic of China
| | | | | | | | | |
Collapse
|
413
|
Hatzikirou H, Alfonso JCL, Mühle S, Stern C, Weiss S, Meyer-Hermann M. Cancer therapeutic potential of combinatorial immuno- and vasomodulatory interventions. J R Soc Interface 2016; 12:rsif.2015.0439. [PMID: 26510827 DOI: 10.1098/rsif.2015.0439] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/23/2023] Open
Abstract
Currently, most of the basic mechanisms governing tumour-immune system interactions, in combination with modulations of tumour-associated vasculature, are far from being completely understood. Here, we propose a mathematical model of vascularized tumour growth, where the main novelty is the modelling of the interplay between functional tumour vasculature and effector cell recruitment dynamics. Parameters are calibrated on the basis of different in vivo immunocompromised Rag1(-/-) and wild-type (WT) BALB/c murine tumour growth experiments. The model analysis supports that tumour vasculature normalization can be a plausible and effective strategy to treat cancer when combined with appropriate immunostimulations. We find that improved levels of functional tumour vasculature, potentially mediated by normalization or stress alleviation strategies, can provide beneficial outcomes in terms of tumour burden reduction and growth control. Normalization of tumour blood vessels opens a therapeutic window of opportunity to augment the antitumour immune responses, as well as to reduce intratumoral immunosuppression and induced hypoxia due to vascular abnormalities. The potential success of normalizing tumour-associated vasculature closely depends on the effector cell recruitment dynamics and tumour sizes. Furthermore, an arbitrary increase in the initial effector cell concentration does not necessarily imply better tumour control. We evidence the existence of an optimal concentration range of effector cells for tumour shrinkage. Based on these findings, we suggest a theory-driven therapeutic proposal that optimally combines immuno- and vasomodulatory interventions.
Collapse
Affiliation(s)
- H Hatzikirou
- Center for Advancing Electronics, Technische Universität Dresden, 01062 Dresden, Germany Center for Information Services and High Performance Computing, Technische Universität Dresden, 01062 Dresden, Germany Department of Systems Immunology and Braunschweig Integrated Centre of Systems Biology, Helmholtz Center for Infectious Research, Inhoffenstrasse 7, 38124 Braunschweig, Germany
| | - J C L Alfonso
- Center for Advancing Electronics, Technische Universität Dresden, 01062 Dresden, Germany Center for Information Services and High Performance Computing, Technische Universität Dresden, 01062 Dresden, Germany
| | - S Mühle
- Molecular Immunology, Helmholtz Center for Infectious Research, Inhoffenstrasse 7, 38124 Braunschweig, Germany
| | - C Stern
- Molecular Immunology, Helmholtz Center for Infectious Research, Inhoffenstrasse 7, 38124 Braunschweig, Germany
| | - S Weiss
- Molecular Immunology, Helmholtz Center for Infectious Research, Inhoffenstrasse 7, 38124 Braunschweig, Germany Institute of Immunology, Medical School Hannover, Carl-Neuberg-Strasse 1, 30625 Hannover, Germany
| | - M Meyer-Hermann
- Department of Systems Immunology and Braunschweig Integrated Centre of Systems Biology, Helmholtz Center for Infectious Research, Inhoffenstrasse 7, 38124 Braunschweig, Germany Institute for Biochemistry, Biotechnology and Bioinformatics, Technische Universität Braunschweig, 38106 Braunschweig, Germany
| |
Collapse
|
414
|
Wang X, Shen Y, Li S, Lv M, Zhang X, Yang J, Wang F, Yang J. Importance of the interaction between immune cells and tumor vasculature mediated by thalidomide in cancer treatment (Review). Int J Mol Med 2016; 38:1021-9. [PMID: 27599781 DOI: 10.3892/ijmm.2016.2724] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2015] [Accepted: 08/25/2016] [Indexed: 11/05/2022] Open
Abstract
Over the past 60 years, thalidomide has metamorphosized from a drug prescribed to treat morning sickness in pregnant women, which was subsequently found to induce birth defects, into a highly effective therapy for treating leprosy and multiple myeloma. Several mechanisms have been proposed to explain the anticancer effects of thalidomide, including antiangiogenic and immunomodulatory activities. At present, evidence suggests that thalidomide may induce vessel maturation. Vascular normalization may be an effective strategy to enhance cancer immunotherapy. Numerous studies have shown that the tumor infiltrating immune cell subsets are important in regulating the process of tumor angiogenesis. The mechanisms associated with antiangiogenesis and the potent immunomodulatory effects of thalidomide obtained the most support. The studies of the antiangiogenic activity of thalidomide were guided in a novel direction by a hypothesis regarding the vascular normalization of tumors. Hence, thalidomide is effective in cancer treatment due to the interaction between immune cells and tumor vasculature. This mechanism provides new avenues to explore for the treatment of cancer.
Collapse
Affiliation(s)
- Xin Wang
- Department of Medical Oncology, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi 710061, P.R. China
| | - Yanwei Shen
- Department of Medical Oncology, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi 710061, P.R. China
| | - Shuting Li
- Department of Medical Oncology, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi 710061, P.R. China
| | - Meng Lv
- Department of Medical Oncology, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi 710061, P.R. China
| | - Xiaoman Zhang
- Department of Medical Oncology, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi 710061, P.R. China
| | - Jiao Yang
- Department of Medical Oncology, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi 710061, P.R. China
| | - Fan Wang
- Department of Medical Oncology, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi 710061, P.R. China
| | - Jin Yang
- Department of Medical Oncology, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi 710061, P.R. China
| |
Collapse
|
415
|
Kalluri R. The biology and function of fibroblasts in cancer. NATURE REVIEWS. CANCER 2016. [PMID: 27550820 DOI: 10.1038/nrc.2016.73.] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Among all cells, fibroblasts could be considered the cockroaches of the human body. They survive severe stress that is usually lethal to all other cells, and they are the only normal cell type that can be live-cultured from post-mortem and decaying tissue. Their resilient adaptation may reside in their intrinsic survival programmes and cellular plasticity. Cancer is associated with fibroblasts at all stages of disease progression, including metastasis, and they are a considerable component of the general host response to tissue damage caused by cancer cells. Cancer-associated fibroblasts (CAFs) become synthetic machines that produce many different tumour components. CAFs have a role in creating extracellular matrix (ECM) structure and metabolic and immune reprogramming of the tumour microenvironment with an impact on adaptive resistance to chemotherapy. The pleiotropic actions of CAFs on tumour cells are probably reflective of them being a heterogeneous and plastic population with context-dependent influence on cancer.
Collapse
Affiliation(s)
- Raghu Kalluri
- Department of Cancer Biology, Metastasis Research Center, University of Texas MD Anderson Cancer Center, Houston, Texas 77054, USA
| |
Collapse
|
416
|
Abstract
Among all cells, fibroblasts could be considered the cockroaches of the human body. They survive severe stress that is usually lethal to all other cells, and they are the only normal cell type that can be live-cultured from post-mortem and decaying tissue. Their resilient adaptation may reside in their intrinsic survival programmes and cellular plasticity. Cancer is associated with fibroblasts at all stages of disease progression, including metastasis, and they are a considerable component of the general host response to tissue damage caused by cancer cells. Cancer-associated fibroblasts (CAFs) become synthetic machines that produce many different tumour components. CAFs have a role in creating extracellular matrix (ECM) structure and metabolic and immune reprogramming of the tumour microenvironment with an impact on adaptive resistance to chemotherapy. The pleiotropic actions of CAFs on tumour cells are probably reflective of them being a heterogeneous and plastic population with context-dependent influence on cancer.
Collapse
Affiliation(s)
- Raghu Kalluri
- Department of Cancer Biology, Metastasis Research Center, University of Texas MD Anderson Cancer Center, Houston, Texas 77054, USA
| |
Collapse
|
417
|
Reardon DA, Gilbert MR, Wick W, Liau L. Immunotherapy for neuro-oncology: the critical rationale for combinatorial therapy. Neuro Oncol 2016; 17 Suppl 7:vii32-vii40. [PMID: 26516225 DOI: 10.1093/neuonc/nov178] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
A successful therapeutic paradigm established historically in oncology involves combining agents with potentially complementary mechanisms of antitumor activity into rationally designed regimens. For example, cocktails of cytotoxic agents, which were carefully designed based on mechanisms of action, dose, and scheduling considerations, have led to dramatic improvements in survival including cures for childhood leukemia, Hodgkin's lymphoma, and several other complex cancers. Outcome for glioblastoma, the most common primary malignant CNS cancer, has been more modest, but nonetheless our current standard of care derives from confirmation that combination therapy surpasses single modality therapy. Immunotherapy has recently come of age for medical oncology with exciting therapeutic benefits achieved by several types of agents including vaccines, adoptive T cells, and immune checkpoint inhibitors against several types of cancers. Nonetheless, most benefits are relatively short, while others are durable but are limited to a minority of treated patients. Critical factors limiting efficacy of immunotherapeutics include insufficient immunogenicity and/or inadequate ability to overcome immunosuppressive factors exploited by tumors. The paradigm of rationally designed combinatorial regimens, originally established by cytotoxic therapy for oncology, may also prove relevant for immunotherapy. Realization of the true therapeutic potential of immunotherapy for medical oncology and neuro-oncology patients may require development of combinatorial regimens that optimize immunogenicity and target tumor adaptive immunosuppressive factors.
Collapse
Affiliation(s)
- David A Reardon
- Center of Neuro-Oncology, Dana-Farber/Brigham and Women's Cancer Center, Boston, Massachusetts (D.A.R.); Department of Medical Oncology, Dana-Farber/Brigham and Women's Cancer Center, Boston, Massachusetts (D.A.R.); Neurology Clinic and National Center for Cancer Research, National Cancer Institute, Bethesda, Maryland (M.R.G.); Neurology Clinic and National Center for Tumor Diseases, University of Heidelberg and German Consortium for Translational Cancer Research (DKTK), German Cancer Research Center (DKFZ), Heidelberg, Germany (W.W); Brain Tumor Program, Department of Neurosurgery, University of California Los Angeles, David Geffen School of Medicine at UCLA, Los Angeles, California (L.L.)
| | - Mark R Gilbert
- Center of Neuro-Oncology, Dana-Farber/Brigham and Women's Cancer Center, Boston, Massachusetts (D.A.R.); Department of Medical Oncology, Dana-Farber/Brigham and Women's Cancer Center, Boston, Massachusetts (D.A.R.); Neurology Clinic and National Center for Cancer Research, National Cancer Institute, Bethesda, Maryland (M.R.G.); Neurology Clinic and National Center for Tumor Diseases, University of Heidelberg and German Consortium for Translational Cancer Research (DKTK), German Cancer Research Center (DKFZ), Heidelberg, Germany (W.W); Brain Tumor Program, Department of Neurosurgery, University of California Los Angeles, David Geffen School of Medicine at UCLA, Los Angeles, California (L.L.)
| | - Wolfgang Wick
- Center of Neuro-Oncology, Dana-Farber/Brigham and Women's Cancer Center, Boston, Massachusetts (D.A.R.); Department of Medical Oncology, Dana-Farber/Brigham and Women's Cancer Center, Boston, Massachusetts (D.A.R.); Neurology Clinic and National Center for Cancer Research, National Cancer Institute, Bethesda, Maryland (M.R.G.); Neurology Clinic and National Center for Tumor Diseases, University of Heidelberg and German Consortium for Translational Cancer Research (DKTK), German Cancer Research Center (DKFZ), Heidelberg, Germany (W.W); Brain Tumor Program, Department of Neurosurgery, University of California Los Angeles, David Geffen School of Medicine at UCLA, Los Angeles, California (L.L.)
| | - Linda Liau
- Center of Neuro-Oncology, Dana-Farber/Brigham and Women's Cancer Center, Boston, Massachusetts (D.A.R.); Department of Medical Oncology, Dana-Farber/Brigham and Women's Cancer Center, Boston, Massachusetts (D.A.R.); Neurology Clinic and National Center for Cancer Research, National Cancer Institute, Bethesda, Maryland (M.R.G.); Neurology Clinic and National Center for Tumor Diseases, University of Heidelberg and German Consortium for Translational Cancer Research (DKTK), German Cancer Research Center (DKFZ), Heidelberg, Germany (W.W); Brain Tumor Program, Department of Neurosurgery, University of California Los Angeles, David Geffen School of Medicine at UCLA, Los Angeles, California (L.L.)
| |
Collapse
|
418
|
Falcon BL, Chintharlapalli S, Uhlik MT, Pytowski B. Antagonist antibodies to vascular endothelial growth factor receptor 2 (VEGFR-2) as anti-angiogenic agents. Pharmacol Ther 2016; 164:204-25. [PMID: 27288725 DOI: 10.1016/j.pharmthera.2016.06.001] [Citation(s) in RCA: 96] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Interaction of numerous signaling pathways in endothelial and mesangial cells results in exquisite control of the process of physiological angiogenesis, with a central role played by vascular endothelial growth factor receptor 2 (VEGFR-2) and its cognate ligands. However, deregulated angiogenesis participates in numerous pathological processes. Excessive activation of VEGFR-2 has been found to mediate tissue-damaging vascular changes as well as the induction of blood vessel expansion to support the growth of solid tumors. Consequently, therapeutic intervention aimed at inhibiting the VEGFR-2 pathway has become a mainstay of treatment in cancer and retinal diseases. In this review, we introduce the concepts of physiological and pathological angiogenesis, the crucial role played by the VEGFR-2 pathway in these processes, and the various inhibitors of its activity that have entered the clinical practice. We primarily focus on the development of ramucirumab, the antagonist monoclonal antibody (mAb) that inhibits VEGFR-2 and has recently been approved for use in patients with gastric, colorectal, and lung cancers. We examine in-depth the pre-clinical studies using DC101, the mAb to mouse VEGFR-2, which provided a conceptual foundation for the role of VEGFR-2 in physiological and pathological angiogenesis. Finally, we discuss further clinical development of ramucirumab and the future of targeting the VEGF pathway for the treatment of cancer.
Collapse
|
419
|
Vago R, Collico V, Zuppone S, Prosperi D, Colombo M. Nanoparticle-mediated delivery of suicide genes in cancer therapy. Pharmacol Res 2016; 111:619-641. [PMID: 27436147 DOI: 10.1016/j.phrs.2016.07.007] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/15/2016] [Revised: 06/27/2016] [Accepted: 07/05/2016] [Indexed: 02/06/2023]
Abstract
Conventional chemotherapeutics have been employed in cancer treatment for decades due to their efficacy in killing the malignant cells, but the other side of the coin showed off-target effects, onset of drug resistance and recurrences. To overcome these limitations, different approaches have been investigated and suicide gene therapy has emerged as a promising alternative. This approach consists in the introduction of genetic materials into cancerous cells or the surrounding tissue to cause cell death or retard the growth of the tumor mass. Despite promising results obtained both in vitro and in vivo, this innovative approach has been limited, for long time, to the treatment of localized tumors, due to the suboptimal efficiency in introducing suicide genes into cancer cells. Nanoparticles represent a valuable non-viral delivery system to protect drugs in the bloodstream, to improve biodistribution, and to limit side effects by achieving target selectivity through surface ligands. In this scenario, the real potential of suicide genes can be translated into clinically viable treatments for patients. In the present review, we summarize the recent advances of inorganic nanoparticles as non-viral vectors in terms of therapeutic efficacy, targeting capacity and safety issues. We describe the main suicide genes currently used in therapy, with particular emphasis on toxin-encoding genes of bacterial and plant origin. In addition, we discuss the relevance of molecular targeting and tumor-restricted expression to improve treatment specificity to cancer tissue. Finally, we analyze the main clinical applications, limitations and future perspectives of suicide gene therapy.
Collapse
Affiliation(s)
- Riccardo Vago
- Università Vita-Salute San Raffaele, Milano, I-20132, Italy; Istituto di Ricerca Urologica, Divisione di Oncologia Sperimentale, IRCCS Ospedale San Raffaele, Via Olgettina 60, 20132, Milan, Italy
| | - Veronica Collico
- Università degli Studi di Milano-Bicocca, NanoBioLab, Dipartimento di Biotecnologie e Bioscienze, Piazza Della Scienza 2, 20126 Milan, Italy
| | - Stefania Zuppone
- Università degli Studi di Milano-Bicocca, NanoBioLab, Dipartimento di Biotecnologie e Bioscienze, Piazza Della Scienza 2, 20126 Milan, Italy; Istituto di Ricerca Urologica, Divisione di Oncologia Sperimentale, IRCCS Ospedale San Raffaele, Via Olgettina 60, 20132, Milan, Italy
| | - Davide Prosperi
- Università degli Studi di Milano-Bicocca, NanoBioLab, Dipartimento di Biotecnologie e Bioscienze, Piazza Della Scienza 2, 20126 Milan, Italy
| | - Miriam Colombo
- Università degli Studi di Milano-Bicocca, NanoBioLab, Dipartimento di Biotecnologie e Bioscienze, Piazza Della Scienza 2, 20126 Milan, Italy.
| |
Collapse
|
420
|
Di N, Mao N, Cheng W, Pang H, Ren Y, Wang N, Liu X, Wang B. Blood oxygenation level-dependent magnetic resonance imaging during carbogen breathing: differentiation between prostate cancer and benign prostate hyperplasia and correlation with vessel maturity. Onco Targets Ther 2016; 9:4143-50. [PMID: 27462169 PMCID: PMC4939976 DOI: 10.2147/ott.s105480] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/23/2023] Open
Abstract
OBJECTIVE The aim of this study was to investigate whether the blood oxygenation level-dependent (BOLD) contrast magnetic resonance imaging (MRI) can evaluate tumor maturity and preoperatively differentiate prostate cancer (PCa) from benign prostate hyperplasia (BPH). PATIENTS AND METHODS BOLD MRI based on transverse relaxation time*-weighted echo planar imaging was performed to assess PCa (19) and BPH (22) responses to carbogen (95% O2 and 5% CO2). The average signal values of PCa and BPH before and after carbogen breathing and the relative increased signal values were computed, respectively. The endothelial-cell marker, CD31, and the pericyte marker, α-smooth muscle actin (mature vessels), were detected with immunofluorescence, and were assessed by microvessel density (MVD) and microvessel pericyte density (MPD). The microvessel pericyte coverage index (MPI) was used to evaluate the degree of vascular maturity. The changed signal from BOLD MRI was correlated with MVD, MPD, and MPI. RESULTS After inhaling carbogen, both PCa and BPH showed an increased signal, but a lower slope was found in PCa than that in BPH (P<0.05). PCa had a higher MPD and MVD but a lower MPI than BPH. The increased signal intensity was positively correlated with MPI in PCa and that in BPH (r=0.616, P=0.011; r=0.658, P=0.002); however, there was no correlation between the increased signal intensity and MPD or MVD in PCa than that in BPH (P>0.05). CONCLUSION Our results confirmed that the increased signal values induced by BOLD MRI well differentiated PCa from BPH and had a positive correlation with vessel maturity in both of them. BOLD MRI can be utilized as a surrogate marker for the noninvasive assessment of the degree of vessel maturity.
Collapse
Affiliation(s)
- Ningning Di
- Department of Radiology, Affiliated Huashan Hospital of Fudan University, Shanghai; Department of Radiology, Binzhou Medical University Affiliated Hospital, Binzhou
| | - Ning Mao
- Department of Radiology, Yantai Yuhangding Hospital, Yantai
| | - Wenna Cheng
- Department of Pharmacy, Binzhou Medical University Affiliated Hospital, Binzhou
| | - Haopeng Pang
- Department of Radiology, Affiliated Huashan Hospital of Fudan University, Shanghai
| | - Yan Ren
- Department of Radiology, Affiliated Huashan Hospital of Fudan University, Shanghai
| | - Ning Wang
- Department of Radiology, Binzhou Medical University Affiliated Hospital, Binzhou
| | - Xinjiang Liu
- Department of Radiology, Binzhou Medical University Affiliated Hospital, Binzhou
| | - Bin Wang
- Department of Medical Imaging and Nuclear Medicine, Binzhou Medical University, Yantai, People's Republic of China
| |
Collapse
|
421
|
Yang YC, Chiang CS. Challenges of Using High-Dose Fractionation Radiotherapy in Combination Therapy. Front Oncol 2016; 6:165. [PMID: 27446811 PMCID: PMC4927577 DOI: 10.3389/fonc.2016.00165] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2015] [Accepted: 06/20/2016] [Indexed: 01/07/2023] Open
Abstract
Radiotherapy is crucial and substantially contributes to multimodal cancer treatment. The combination of conventional fractionation radiotherapy (CFRT) and systemic therapy has been established as the standard treatment for many cancer types. With advances in linear accelerators and image-guided techniques, high-dose fractionation radiotherapy (HFRT) is increasingly introduced in cancer centers. Clinicians are currently integrating HFRT into multimodality treatment. The shift from CFRT to HFRT reveals different effects on the tumor microenvironment and responses, particularly the immune response. Furthermore, the combination of HFRT and drugs yields different results in different types of tumors or using different treatment schemes. We have reviewed clinical trials and preclinical evidence on the combination of HFRT with drugs, such as chemotherapy, targeted therapy, and immune therapy. Notably, HFRT apparently enhances tumor cell killing and antigen presentation, thus providing opportunities and challenges in treating cancer.
Collapse
Affiliation(s)
- Ying-Chieh Yang
- Biomedical Engineering and Environmental Sciences, National Tsing Hua University, Hsinchu City, Taiwan
- Radiation Oncology, National Taiwan University Hospital Hsin-Chu Branch, Hsinchu City, Taiwan
| | - Chi-Shiun Chiang
- Biomedical Engineering and Environmental Sciences, National Tsing Hua University, Hsinchu City, Taiwan
| |
Collapse
|
422
|
Beatty GL, O'Hara M. Chimeric antigen receptor-modified T cells for the treatment of solid tumors: Defining the challenges and next steps. Pharmacol Ther 2016; 166:30-9. [PMID: 27373504 DOI: 10.1016/j.pharmthera.2016.06.010] [Citation(s) in RCA: 96] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/08/2016] [Indexed: 01/07/2023]
Abstract
Chimeric antigen receptor (CAR) T cell therapy has shown promise in CD19 expressing hematologic malignancies, but how to translate this success to solid malignancies remains elusive. Effective translation of CAR T cells to solid tumors will require an understanding of potential therapeutic barriers, including factors that regulate CAR T cells expansion, persistence, trafficking, and fate within tumors. Herein, we describe the current state of CAR T cells in solid tumors; define key barriers to CAR T cell efficacy and mechanisms underlying these barriers, outline potential avenues for overcoming these therapeutic obstacles, and discuss the future of translating CAR T cells for the treatment of patients with solid malignancies.
Collapse
Affiliation(s)
- Gregory L Beatty
- Abramson Cancer Center, University of Pennsylvania, Philadelphia, PA, USA; Division of Hematology-Oncology, Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA.
| | - Mark O'Hara
- Abramson Cancer Center, University of Pennsylvania, Philadelphia, PA, USA; Division of Hematology-Oncology, Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| |
Collapse
|
423
|
Mukaida N, Sasaki S. Fibroblasts, an inconspicuous but essential player in colon cancer development and progression. World J Gastroenterol 2016; 22:5301-5316. [PMID: 27340347 PMCID: PMC4910652 DOI: 10.3748/wjg.v22.i23.5301] [Citation(s) in RCA: 42] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/16/2016] [Revised: 04/22/2016] [Accepted: 05/21/2016] [Indexed: 02/06/2023] Open
Abstract
Tumor microenvironments have a crucial role in cancer initiation and progression, and share many molecular and pathological features with wound healing process. Unless treated, tumors, however, do not heal in contrast to wounds that heal within a limited time framework. Wounds heal in coordination of a myriad of types of cells, particularly endothelial cells, leukocytes, and fibroblasts. Similar sets of cells also contribute to cancer initiation and progression, and as a consequence, anti-cancer treatment strategies have been proposed and tested by targeting endothelial cells and/or leukocytes. Compared with endothelial cells and leukocytes, less attention has been paid to the roles of cancer-associated fibroblasts (CAFs), fibroblasts present in tumor tissues, because their heterogeneity hinders the elucidation on them at cellular and molecular levels. Here, we will discuss the origin of CAFs and their crucial roles in cancer initiation and progression, and the possibility to develop a novel type of anti-cancer treatment by manipulating the migration and functions of CAFs.
Collapse
|
424
|
Fukuda T, Kamai T, Masuda A, Nukui A, Abe H, Arai K, Yoshida KI. Higher preoperative serum levels of PD-L1 and B7-H4 are associated with invasive and metastatic potential and predictable for poor response to VEGF-targeted therapy and unfavorable prognosis of renal cell carcinoma. Cancer Med 2016; 5:1810-20. [PMID: 27292320 PMCID: PMC4971909 DOI: 10.1002/cam4.754] [Citation(s) in RCA: 46] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2016] [Revised: 02/29/2016] [Accepted: 04/05/2016] [Indexed: 12/19/2022] Open
Abstract
Renal cell carcinoma (RCC) is an immunogenic and proangiogenic cancer. Although antivascular endothelial growth factor (VEGF) therapies achieve impressive responses in some patients, many tumors eventually develop resistance to such therapy. The B7 family molecules such as CTLA‐4, PD‐1, and PD‐L1 are pivotal players in immune checkpoints that positively or negatively regulate various immune responses. Recently, immunotherapy based on blocking immune checkpoints with anti‐CTLA4, anti‐PD‐1, or anti‐PD‐L1 antibodies has been proposed as a potential new approach to the treatment of metastatic RCC. Higher expression of PD‐L1 and B7‐H4 in the tumors is associated with a poor prognosis in RCCs, however, the clinical impact of serum levels of B7 family molecules has not been elucidated in patients with metastatic RCCs receiving VEGF‐targeted agents. We assessed the preoperative serum levels of B7 family molecules, including CD80, CD86, PD‐1, PD‐L1, B7‐H3, B7‐H4, and CTLA‐4, and CD28 in RCC patients, and determined their relations with various clinicopathological characteristics. Elevated preoperative serum levels of PD‐L1 and B7‐H4 were correlated with less differentiated tumors, higher invasive and metastatic potential, a worse response to anti‐VEGF therapy, and shorter overall survival. These findings suggested that investigating preoperative serum levels of PD‐L1 and B7‐H4 might not only be useful to assess the biological aggressiveness of RCCs, but also to predict the efficacy of anti‐VEGF therapy and the eventual prognosis, indicating the future design of clinical trials of therapies targeting immune checkpoint in advanced RCCs.
Collapse
Affiliation(s)
- Takehiko Fukuda
- Department of Urology, Dokkyo Medical University, Tochigi, Japan
| | - Takao Kamai
- Department of Urology, Dokkyo Medical University, Tochigi, Japan
| | - Akinori Masuda
- Dialysis center, Dokkyo Medical University Koshigaya Hospital, Saitama, Japan
| | - Akinori Nukui
- Department of Urology, Nasu Red Cross Hospital, Tochigi, Japan
| | - Hideyuki Abe
- Department of Urology, Dokkyo Medical University, Tochigi, Japan
| | - Kyoko Arai
- Department of Urology, Dokkyo Medical University, Tochigi, Japan
| | | |
Collapse
|
425
|
Wang Z, Kerketta R, Chuang YL, Dogra P, Butner JD, Brocato TA, Day A, Xu R, Shen H, Simbawa E, AL-Fhaid AS, Mahmoud SR, Curley SA, Ferrari M, Koay EJ, Cristini V. Theory and Experimental Validation of a Spatio-temporal Model of Chemotherapy Transport to Enhance Tumor Cell Kill. PLoS Comput Biol 2016; 12:e1004969. [PMID: 27286441 PMCID: PMC4902302 DOI: 10.1371/journal.pcbi.1004969] [Citation(s) in RCA: 42] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2015] [Accepted: 05/09/2016] [Indexed: 12/14/2022] Open
Abstract
It has been hypothesized that continuously releasing drug molecules into the tumor over an extended period of time may significantly improve the chemotherapeutic efficacy by overcoming physical transport limitations of conventional bolus drug treatment. In this paper, we present a generalized space- and time-dependent mathematical model of drug transport and drug-cell interactions to quantitatively formulate this hypothesis. Model parameters describe: perfusion and tissue architecture (blood volume fraction and blood vessel radius); diffusion penetration distance of drug (i.e., a function of tissue compactness and drug uptake rates by tumor cells); and cell death rates (as function of history of drug uptake). We performed preliminary testing and validation of the mathematical model using in vivo experiments with different drug delivery methods on a breast cancer mouse model. Experimental data demonstrated a 3-fold increase in response using nano-vectored drug vs. free drug delivery, in excellent quantitative agreement with the model predictions. Our model results implicate that therapeutically targeting blood volume fraction, e.g., through vascular normalization, would achieve a better outcome due to enhanced drug delivery.
Collapse
Affiliation(s)
- Zhihui Wang
- Department of NanoMedicine and Biomedical Engineering, University of Texas Medical School at Houston, Houston, Texas, United States of America
- Brown Foundation Institute of Molecular Medicine, University of Texas Medical School at Houston, Houston, Texas, United States of America
- Department of Imaging Physics, University of Texas MD Anderson Cancer Center, Houston, Texas, United States of America
| | - Romica Kerketta
- Department of Pathology, University of New Mexico, Albuquerque, New Mexico, United States of America
| | - Yao-Li Chuang
- Department of Mathematics, California State University, Northridge, California, United States of America
| | - Prashant Dogra
- Department of Pathology, University of New Mexico, Albuquerque, New Mexico, United States of America
| | - Joseph D. Butner
- Department of Chemical and Biological Engineering and Center for Biomedical Engineering, University of New Mexico, Albuquerque, New Mexico, United States of America
| | - Terisse A. Brocato
- Department of Chemical and Biological Engineering and Center for Biomedical Engineering, University of New Mexico, Albuquerque, New Mexico, United States of America
| | - Armin Day
- Department of Pathology, University of New Mexico, Albuquerque, New Mexico, United States of America
| | - Rong Xu
- Department of Nanomedicine, Methodist Hospital Research Institute, Houston, Texas, United States of America
| | - Haifa Shen
- Department of Nanomedicine, Methodist Hospital Research Institute, Houston, Texas, United States of America
| | - Eman Simbawa
- Department of Mathematics, Faculty of Science, King Abdulaziz University, Jeddah, Saudi Arabia
| | - A. S. AL-Fhaid
- Department of Mathematics, Faculty of Science, King Abdulaziz University, Jeddah, Saudi Arabia
| | - S. R. Mahmoud
- Department of Mathematics, Faculty of Science, King Abdulaziz University, Jeddah, Saudi Arabia
| | - Steven A. Curley
- Michael E. DeBakey Department of Surgery, Baylor College of Medicine, Houston, Texas, United States of America
| | - Mauro Ferrari
- Department of Nanomedicine, Methodist Hospital Research Institute, Houston, Texas, United States of America
| | - Eugene J. Koay
- Department of Radiation Oncology, University of Texas MD Anderson Cancer Center, Houston, Texas, United States of America
- * E-mail: (EJK); (VC)
| | - Vittorio Cristini
- Department of NanoMedicine and Biomedical Engineering, University of Texas Medical School at Houston, Houston, Texas, United States of America
- Brown Foundation Institute of Molecular Medicine, University of Texas Medical School at Houston, Houston, Texas, United States of America
- Department of Imaging Physics, University of Texas MD Anderson Cancer Center, Houston, Texas, United States of America
- Department of Mathematics, Faculty of Science, King Abdulaziz University, Jeddah, Saudi Arabia
- * E-mail: (EJK); (VC)
| |
Collapse
|
426
|
Moving Immune Checkpoint Blockade in Thoracic Tumors beyond NSCLC. J Thorac Oncol 2016; 11:1819-1836. [PMID: 27288978 DOI: 10.1016/j.jtho.2016.05.027] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2016] [Revised: 05/24/2016] [Accepted: 05/28/2016] [Indexed: 02/07/2023]
Abstract
SCLC and malignant pleural mesothelioma (MPM) are historically characterized by a disappointing lack of significant therapeutic breakthroughs for novel agents, and both malignancies represent true unmet medical needs. Given the promising results of anti-cytotoxic T-lymphocyte associated protein-4 and anti-programmed cell death-1/programmed death ligand-1 antibodies in the treatment of advanced NSCLCs, these immune checkpoint inhibitors are now also under investigation in SCLC and MPM, as well as in thymic epithelial tumors (TETs). Here, we review the biological and clinical rationale for immune checkpoint inhibition in SCLC, MPM, and TETs and present preliminary clinical results with available antibodies. Immunotherapeutic perspectives for these malignancies in terms of novel agents currently under evaluation or anticipated in the near future are also discussed. Current immune checkpoint blockers targeting cytotoxic T-lymphocyte associated protein-4 and the programmed cell death-1/programmed death ligand-1 axis, administered alone or in combination and as multimodality treatment, are likely to be a valuable addition to the therapeutic array for managing SCLC and MPM; studies in TETs, which are currently in their infancy, are merited. Close attention to potential toxicities will be important to the success of such strategies in these settings.
Collapse
|
427
|
Du Four S, Maenhout SK, Benteyn D, De Keersmaecker B, Duerinck J, Thielemans K, Neyns B, Aerts JL. Disease progression in recurrent glioblastoma patients treated with the VEGFR inhibitor axitinib is associated with increased regulatory T cell numbers and T cell exhaustion. Cancer Immunol Immunother 2016; 65:727-40. [PMID: 27098427 PMCID: PMC11029796 DOI: 10.1007/s00262-016-1836-3] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2015] [Accepted: 04/01/2016] [Indexed: 01/03/2023]
Abstract
BACKGROUND Recurrent glioblastoma is associated with a poor overall survival. Antiangiogenic therapy results in a high tumor response rate but has limited impact on survival. Immunotherapy has emerged as an efficient treatment modality for some cancers, and preclinical evidence indicates that anti-VEGF(R) therapy can counterbalance the immunosuppressive tumor microenvironment. METHODS We collected peripheral blood mononuclear cells (PBMC) of patients with recurrent glioblastoma treated in a randomized phase II clinical trial comparing the effect of axitinib with axitinib plus lomustine and analyzed the immunophenotype of PBMC, the production of cytokines and expression of inhibitory molecules by circulating T cells. RESULTS PBMC of 18 patients were collected at baseline and at 6 weeks after initiation of study treatment. Axitinib increased the number of naïve CD8(+) T cells and central memory CD4(+) and CD8(+) T cells and reduced the TIM3 expression on CD4(+) and CD8(+) T cells. Patients diagnosed with progressive disease on axitinib had a significantly increased number of regulatory T cells and an increased level of PD-1 expression on CD4(+) and CD8(+) T cells. In addition, reduced numbers of cytokine-producing T cells were found in progressive patients as compared to patients responding to treatment. CONCLUSION Our results suggest that axitinib treatment in patients with recurrent glioblastoma has a favorable impact on immune function. At the time of acquired resistance to axitinib, we documented further enhancement of a preexisting immunosuppression. Further investigations on the role of axitinib as potential combination partner with immunotherapy are necessary.
Collapse
Affiliation(s)
- Stephanie Du Four
- Laboratory of Molecular and Cellular Therapy, Vrije Universiteit Brussel, Laarbeeklaan 103E, 1090, Brussels, Belgium
| | - Sarah K Maenhout
- Laboratory of Molecular and Cellular Therapy, Vrije Universiteit Brussel, Laarbeeklaan 103E, 1090, Brussels, Belgium
| | - Daphné Benteyn
- Laboratory of Molecular and Cellular Therapy, Vrije Universiteit Brussel, Laarbeeklaan 103E, 1090, Brussels, Belgium
| | - Brenda De Keersmaecker
- Laboratory of Molecular and Cellular Therapy, Vrije Universiteit Brussel, Laarbeeklaan 103E, 1090, Brussels, Belgium
| | - Johnny Duerinck
- Department of Neurosurgery, Universitair Ziekenhuis Brussel, Laarbeeklaan 101, 1090, Brussels, Belgium
| | - Kris Thielemans
- Laboratory of Molecular and Cellular Therapy, Vrije Universiteit Brussel, Laarbeeklaan 103E, 1090, Brussels, Belgium
| | - Bart Neyns
- Department of Medical Oncology, Universitair Ziekenhuis Brussel, Laarbeeklaan 101, 1090, Brussels, Belgium
| | - Joeri L Aerts
- Laboratory of Molecular and Cellular Therapy, Vrije Universiteit Brussel, Laarbeeklaan 103E, 1090, Brussels, Belgium.
| |
Collapse
|
428
|
Urup T, Michaelsen SR, Olsen LR, Toft A, Christensen IJ, Grunnet K, Winther O, Broholm H, Kosteljanetz M, Issazadeh-Navikas S, Poulsen HS, Lassen U. Angiotensinogen and HLA class II predict bevacizumab response in recurrent glioblastoma patients. Mol Oncol 2016; 10:1160-8. [PMID: 27262894 DOI: 10.1016/j.molonc.2016.05.005] [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: 03/05/2016] [Revised: 05/01/2016] [Accepted: 05/19/2016] [Indexed: 12/26/2022] Open
Abstract
BACKGROUND Bevacizumab combination therapy is among the most frequently used treatments in recurrent glioblastoma and patients who achieve response to bevacizumab have improved survival as well as quality of life. Accordingly, the aim of this study was to identify predictive biomarkers for bevacizumab response in recurrent glioblastoma patients. METHODS The study included a total of 82 recurrent glioblastoma patients treated with bevacizumab combination therapy whom were both response and biomarker evaluable. Gene expression of tumor tissue was analyzed by using a customized NanoString platform covering 800 genes. Candidate gene predictors associated with response were analyzed by multivariate logistic and Cox regression analysis. RESULTS Two genes were independently associated with response: Low expression of angiotensinogen (2-fold decrease in AGT; OR = 2.44; 95% CI: 1.45-4.17; P = 0.0009) and high expression of a HLA class II gene (2-fold increase in HLA-DQA1; OR = 1.22; 95% CI: 1.01-1.47; P = 0.04). These two genes were included in a model that is able predict response to bevacizumab combination therapy in clinical practice. When stratified for a validated prognostic index, the predictive model for response was significantly associated with improved overall survival. CONCLUSION Two genes (low angiotensinogen and high HLA-class II expression) were predictive for bevacizumab response and were included in a predictive model for response. This model can be used in clinical practice to identify patients who will benefit from bevacizumab combination therapy.
Collapse
Affiliation(s)
- Thomas Urup
- Department of Radiation Biology, The Finsen Center, Rigshospitalet, Blegdamsvej 9, DK-2100 Copenhagen, Denmark.
| | - Signe Regner Michaelsen
- Department of Radiation Biology, The Finsen Center, Rigshospitalet, Blegdamsvej 9, DK-2100 Copenhagen, Denmark
| | - Lars Rønn Olsen
- Center for Biological Sequence Analysis, Department of Systems Biology, Technical University of Denmark, Kemitorvet, Building 208, DK-2800 Lyngby, Denmark; Bioinformatics Centre, Department of Biology and Biotech Research and Innovation Centre (BRIC), University of Copenhagen, DK-2200, Denmark
| | - Anders Toft
- Department of Radiation Biology, The Finsen Center, Rigshospitalet, Blegdamsvej 9, DK-2100 Copenhagen, Denmark
| | - Ib Jarle Christensen
- Department of Gastroenterology, Hvidovre Hospital, Kettegård Allé 30, DK-2650 Hvidovre, Denmark
| | - Kirsten Grunnet
- Department of Radiation Biology, The Finsen Center, Rigshospitalet, Blegdamsvej 9, DK-2100 Copenhagen, Denmark
| | - Ole Winther
- Bioinformatics Centre, Department of Biology and Biotech Research and Innovation Centre (BRIC), University of Copenhagen, DK-2200, Denmark
| | - Helle Broholm
- Department of Neuropathology, Center of Diagnostic Investigation, Rigshospitalet, Blegdamsvej 9, DK-2100 Copenhagen, Denmark
| | - Michael Kosteljanetz
- Department of Neurosurgery, The Neurocenter, Rigshospitalet, Blegdamsvej 9, DK-2100 Copenhagen, Denmark
| | | | - Hans Skovgaard Poulsen
- Department of Radiation Biology, The Finsen Center, Rigshospitalet, Blegdamsvej 9, DK-2100 Copenhagen, Denmark; Department of Oncology, The Finsen Center, Rigshospitalet, Blegdamsvej 9, DK-2100 Copenhagen, Denmark
| | - Ulrik Lassen
- Department of Radiation Biology, The Finsen Center, Rigshospitalet, Blegdamsvej 9, DK-2100 Copenhagen, Denmark; Department of Oncology, The Finsen Center, Rigshospitalet, Blegdamsvej 9, DK-2100 Copenhagen, Denmark; Phase I Unit, Finsencenter, Rigshospitalet, Blegdamsvej 9, DK-2100 Copenhagen, Denmark
| |
Collapse
|
429
|
Targeted Therapy and Checkpoint Immunotherapy Combinations for the Treatment of Cancer. Trends Immunol 2016; 37:462-476. [PMID: 27216414 DOI: 10.1016/j.it.2016.04.010] [Citation(s) in RCA: 215] [Impact Index Per Article: 23.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2016] [Revised: 04/27/2016] [Accepted: 04/29/2016] [Indexed: 12/11/2022]
Abstract
Many advances in the treatment of cancer have been driven by the development of targeted therapies that inhibit oncogenic signaling pathways and tumor-associated angiogenesis, as well as by the recent development of therapies that activate a patient's immune system to unleash antitumor immunity. Some targeted therapies can have effects on host immune responses, in addition to their effects on tumor biology. These immune-modulating effects, such as increasing tumor antigenicity or promoting intratumoral T cell infiltration, provide a rationale for combining these targeted therapies with immunotherapies. Here, we discuss the immune-modulating effects of targeted therapies against the MAPK and VEGF signaling pathways, and how they may synergize with immunomodulatory antibodies that target PD1/PDL1 and CTLA4. We critically examine the rationale in support of these combinations in light of the current understanding of the underlying mechanisms of action of these therapies. We also discuss the available preclinical and clinical data for these combination approaches and their implications regarding mechanisms of action. Insights from these studies provide a framework for considering additional combinations of targeted therapies and immunotherapies for the treatment of cancer.
Collapse
|
430
|
Leuci V, Maione F, Rotolo R, Giraudo E, Sassi F, Migliardi G, Todorovic M, Gammaitoni L, Mesiano G, Giraudo L, Luraghi P, Leone F, Bussolino F, Grignani G, Aglietta M, Trusolino L, Bertotti A, Sangiolo D. Lenalidomide normalizes tumor vessels in colorectal cancer improving chemotherapy activity. J Transl Med 2016; 14:119. [PMID: 27149858 PMCID: PMC4857418 DOI: 10.1186/s12967-016-0872-2] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2015] [Accepted: 04/20/2016] [Indexed: 12/19/2022] Open
Abstract
Background Angiogenesis inhibition is a promising approach for treating metastatic colorectal cancer (mCRC). Recent evidences support the seemingly counterintuitive ability of certain antiangiogenic drugs to promote normalization of residual tumor vessels with important clinical implications. Lenalidomide is an oral drug with immune-modulatory and anti-angiogenic activity against selected hematologic malignancies but as yet little is known regarding its effectiveness for solid tumors. The aim of this study was to determine whether lenalidomide can normalize colorectal cancer neo-vessels in vivo, thus reducing tumor hypoxia and improving the benefit of chemotherapy. Methods We set up a tumorgraft model with NOD/SCID mice implanted with a patient-derived colorectal cancer liver metastasis. The mice were treated with oral lenalidomide (50 mg/Kg/day for 28 days), intraperitoneal 5-fluorouracil (5FU) (20 mg/Kg twice weekly for 3 weeks), combination (combo) of lenalidomide and 5FU or irrelevant vehicle. We assessed tumor vessel density (CD146), pericyte coverage (NG2; alphaSMA), in vivo perfusion capability of residual vessels (lectin distribution essay), hypoxic areas (HP2-100 Hypoxyprobe) and antitumor activity in vivo and in vitro. Results Treatment with lenalidomide reduced tumor vessel density (p = 0.0001) and enhanced mature pericyte coverage of residual vessels (p = 0.002). Perfusion capability of tumor vessels was enhanced in mice treated with lenalidomide compared to controls (p = 0.004). Accordingly, lenalidomide reduced hypoxic tumor areas (p = 0.002) and enhanced the antitumor activity of 5FU in vivo. The combo treatment delayed tumor growth (p = 0.01) and significantly reduced the Ki67 index (p = 0.0002). Lenalidomide alone did not demonstrate antitumor activity compared to untreated controls in vivo or against 4 different mCRC cell lines in vitro. Conclusions We provide the first evidence of tumor vessel normalization and hypoxia reduction induced by lenalidomide in mCRC in vivo. This effect, seemingly counterintuitive for an antiangiogenic compound, translates into indirect antitumor activity thus enhancing the therapeutic index of chemotherapy. Our findings suggest that further research should be carried out on synergism between lenalidomide and conventional therapies for treating solid tumors that might benefit from tumor vasculature normalization.
Collapse
Affiliation(s)
- V Leuci
- Department of Oncology, University of Torino, Turin, Italy.,Laboratory of Medical Oncology-Experimental Cell Therapy, Candiolo Cancer Institute-FPO- IRCCS, Candiolo, Turin, Italy
| | - F Maione
- Laboratory of Transgenic Mouse Models, Candiolo Cancer Institute-FPO- IRCCS, Candiolo, Turin, Italy
| | - R Rotolo
- Department of Oncology, University of Torino, Turin, Italy.,Laboratory of Medical Oncology-Experimental Cell Therapy, Candiolo Cancer Institute-FPO- IRCCS, Candiolo, Turin, Italy
| | - E Giraudo
- Laboratory of Transgenic Mouse Models, Candiolo Cancer Institute-FPO- IRCCS, Candiolo, Turin, Italy.,Department of Science and Drug Technology, University of Torino, Turin, Italy
| | - F Sassi
- Laboratory of Translational Cancer Medicine, Candiolo Cancer Institute-FPO- IRCCS, Candiolo, Turin, Italy
| | - G Migliardi
- Laboratory of Translational Cancer Medicine, Candiolo Cancer Institute-FPO- IRCCS, Candiolo, Turin, Italy
| | - M Todorovic
- Laboratory of Medical Oncology-Experimental Cell Therapy, Candiolo Cancer Institute-FPO- IRCCS, Candiolo, Turin, Italy
| | - L Gammaitoni
- Laboratory of Medical Oncology-Experimental Cell Therapy, Candiolo Cancer Institute-FPO- IRCCS, Candiolo, Turin, Italy
| | - G Mesiano
- Laboratory of Medical Oncology-Experimental Cell Therapy, Candiolo Cancer Institute-FPO- IRCCS, Candiolo, Turin, Italy
| | - L Giraudo
- Laboratory of Medical Oncology-Experimental Cell Therapy, Candiolo Cancer Institute-FPO- IRCCS, Candiolo, Turin, Italy
| | - P Luraghi
- Laboratory of Cancer Stem Cell Research, Candiolo Cancer Institute-FPO- IRCCS, Candiolo, Turin, Italy
| | - F Leone
- Department of Oncology, University of Torino, Turin, Italy.,Division and Laboratory of Medical Oncology, Candiolo Cancer Institute-FPO- IRCCS, Candiolo, Turin, Italy
| | - F Bussolino
- Department of Oncology, University of Torino, Turin, Italy.,Laboratory of Vascular Oncology, Candiolo Cancer Institute, Candiolo, Turin, Italy
| | - G Grignani
- Division and Laboratory of Medical Oncology, Candiolo Cancer Institute-FPO- IRCCS, Candiolo, Turin, Italy
| | - M Aglietta
- Department of Oncology, University of Torino, Turin, Italy.,Division and Laboratory of Medical Oncology, Candiolo Cancer Institute-FPO- IRCCS, Candiolo, Turin, Italy
| | - L Trusolino
- Department of Oncology, University of Torino, Turin, Italy.,Laboratory of Translational Cancer Medicine, Candiolo Cancer Institute-FPO- IRCCS, Candiolo, Turin, Italy
| | - A Bertotti
- Department of Oncology, University of Torino, Turin, Italy.,Laboratory of Translational Cancer Medicine, Candiolo Cancer Institute-FPO- IRCCS, Candiolo, Turin, Italy
| | - D Sangiolo
- Department of Oncology, University of Torino, Turin, Italy. .,Laboratory of Medical Oncology-Experimental Cell Therapy, Candiolo Cancer Institute-FPO- IRCCS, Candiolo, Turin, Italy.
| |
Collapse
|
431
|
Tada K, Kitano S, Shoji H, Nishimura T, Shimada Y, Nagashima K, Aoki K, Hiraoka N, Honma Y, Iwasa S, Okita N, Takashima A, Kato K, Yamada Y, Katayama N, Boku N, Heike Y, Hamaguchi T. Pretreatment Immune Status Correlates with Progression-Free Survival in Chemotherapy-Treated Metastatic Colorectal Cancer Patients. Cancer Immunol Res 2016; 4:592-9. [PMID: 27197061 DOI: 10.1158/2326-6066.cir-15-0298] [Citation(s) in RCA: 53] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2015] [Accepted: 03/18/2016] [Indexed: 12/29/2022]
Abstract
It remains unclear whether the immunologic status of cells in peripheral blood can be used as a prognostic indicator of response to treatment for patients with unresectable metastatic colorectal cancer (MCRC). We therefore investigated the relationship between the pretreatment immunologic status of 40 patients with MCRC who planned to receive the first-line chemotherapy and their progression-free survival. Twenty-five immune cell subsets, including monocytic myeloid-derived suppressor cells (M-MDSC) and effector memory T cells (TEM), were measured by multicolor-flow cytometry. We divided patients into high and low (above and below the median, respectively) groups based on the median value for each immune cell subset and compared progression-free survival of the two groups. Patients with high M-MDSC, low CD4(+) TEM, or low CD8(+) TEM quantities had significantly shorter progression-free survival (P = 0.004, 0.005, and 0.002, respectively). Patients were classified into two prognostic groups based on numbers of adverse factors; having two or three adverse factors (n = 21, 52.5%) was correlated with significantly shorter progression-free survival compared with none or one (n = 19, 47.5%; P < 0.001). The presence of two or three adverse factors was an independent poor prognostic factor for progression-free survival (HR, 9.2; 95% confidence interval, 2.5-34.2; P < 0.001). These results provide evidence that pretreatment peripheral immune status can inform the outcome of patients with MCRC treated with first-line chemotherapy. Cancer Immunol Res; 4(7); 592-9. ©2016 AACR.
Collapse
Affiliation(s)
- Kohei Tada
- National Cancer Center Hospital, Tokyo, Japan. Department of Hematology and Oncology, Mie University Graduate School of Medicine, Mie, Japan. Immunotherapy and Cell Therapy Service, St. Luke's International Hospital, Tokyo, Japan
| | - Shigehisa Kitano
- Department of Experimental Therapeutics (ex. Phase I trial center), National Cancer Center Hospital, Tokyo, Japan. Exploratory Oncology Research and Clinical Trial Center, Division of Cancer Immunotherapy, National Cancer Center, Tokyo, Japan
| | - Hirokazu Shoji
- Gastrointestinal Medical Oncology Division, National Cancer Center Hospital, Tokyo, Japan
| | - Takashi Nishimura
- Department of Gastrointestinal Medicine, Tokyo Jikei Medical University, Tokyo, Japan
| | - Yasuhiro Shimada
- Gastrointestinal Medical Oncology Division, National Cancer Center Hospital, Tokyo, Japan
| | - Kengo Nagashima
- Department of Global Clinical Research, Graduate School of Medicine, Chiba University, Chiba, Japan
| | - Kazunori Aoki
- Division of Molecular and Cellular Medicine, National Cancer Center Research Institute, Tokyo, Japan
| | - Nobuyoshi Hiraoka
- Division of Molecular Pathology, National Cancer Center Research Institute, Tokyo, Japan
| | - Yoshitaka Honma
- Gastrointestinal Medical Oncology Division, National Cancer Center Hospital, Tokyo, Japan
| | - Satoru Iwasa
- Gastrointestinal Medical Oncology Division, National Cancer Center Hospital, Tokyo, Japan
| | - Natsuko Okita
- Gastrointestinal Medical Oncology Division, National Cancer Center Hospital, Tokyo, Japan
| | - Atsuo Takashima
- Gastrointestinal Medical Oncology Division, National Cancer Center Hospital, Tokyo, Japan
| | - Ken Kato
- Gastrointestinal Medical Oncology Division, National Cancer Center Hospital, Tokyo, Japan
| | - Yasuhide Yamada
- Gastrointestinal Medical Oncology Division, National Cancer Center Hospital, Tokyo, Japan
| | - Naoyuki Katayama
- Department of Hematology and Oncology, Mie University Graduate School of Medicine, Mie, Japan
| | - Narikazu Boku
- Gastrointestinal Medical Oncology Division, National Cancer Center Hospital, Tokyo, Japan
| | - Yuji Heike
- National Cancer Center Hospital, Tokyo, Japan. Immunotherapy and Cell Therapy Service, St. Luke's International Hospital, Tokyo, Japan.
| | - Tetsuya Hamaguchi
- Gastrointestinal Medical Oncology Division, National Cancer Center Hospital, Tokyo, Japan
| |
Collapse
|
432
|
Kirui DK, Ferrari M. Intravital Microscopy Imaging Approaches for Image-Guided Drug Delivery Systems. Curr Drug Targets 2016; 16:528-41. [PMID: 25901526 DOI: 10.2174/1389450116666150330114030] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2014] [Revised: 12/10/2014] [Accepted: 03/13/2015] [Indexed: 12/31/2022]
Abstract
Rapid technical advances in the field of non-linear microscopy have made intravital microscopy a vital pre-clinical tool for research and development of imaging-guided drug delivery systems. The ability to dynamically monitor the fate of macromolecules in live animals provides invaluable information regarding properties of drug carriers (size, charge, and surface coating), physiological, and pathological processes that exist between point-of-injection and the projected of site of delivery, all of which influence delivery and effectiveness of drug delivery systems. In this Review, we highlight how integrating intravital microscopy imaging with experimental designs (in vitro analyses and mathematical modeling) can provide unique information critical in the design of novel disease-relevant drug delivery platforms with improved diagnostic and therapeutic indexes. The Review will provide the reader an overview of the various applications for which intravital microscopy has been used to monitor the delivery of diagnostic and therapeutic agents and discuss some of their potential clinical applications.
Collapse
Affiliation(s)
| | - Mauro Ferrari
- Houston Methodist Research Institute, Department of NanoMedicine, 6670 Bertner Avenue, MS R8-460, Houston, TX 77030, USA.
| |
Collapse
|
433
|
Tiper IV, Temkin SM, Spiegel S, Goldblum SE, Giuntoli RL, Oelke M, Schneck JP, Webb TJ. VEGF Potentiates GD3-Mediated Immunosuppression by Human Ovarian Cancer Cells. Clin Cancer Res 2016; 22:4249-58. [PMID: 27076627 DOI: 10.1158/1078-0432.ccr-15-2518] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2015] [Accepted: 03/29/2016] [Indexed: 12/31/2022]
Abstract
PURPOSE Natural killer T (NKT) cells are important mediators of antitumor immune responses. We have previously shown that ovarian cancers shed the ganglioside GD3, which inhibits NKT-cell activation. Ovarian cancers also secrete high levels of VEGF. In this study, we sought to test the hypothesis that VEGF production by ovarian cancers suppresses NKT-cell-mediated antitumor responses. EXPERIMENTAL DESIGN To investigate the effects of VEGF on CD1d-mediated NKT-cell activation, a conditioned media model was established, wherein the supernatants from ovarian cancer cell lines (OV-CAR-3 and SK-OV-3) were used to treat CD1d-expressing antigen-presenting cells (APC) and cocultured with NKT hybridomas. Ovarian cancer-associated VEGF was inhibited by treatment with bevacizumab and genistein; conditioned medium was collected, and CD1d-mediated NKT-cell responses were assayed by ELISA. RESULTS Ovarian cancer tissue and ascites contain lymphocytic infiltrates, suggesting that immune cells traffic to tumors, but are then inhibited by immunosuppressive molecules within the tumor microenvironment. OV-CAR-3 and SK-OV-3 cell lines produce high levels of VEGF and GD3. Pretreatment of APCs with ascites or conditioned medium from OV-CAR-3 and SK-OV-3 blocked CD1d-mediated NKT-cell activation. Inhibition of VEGF resulted in a concomitant reduction in GD3 levels and restoration of NKT-cell responses. CONCLUSIONS We found that VEGF inhibition restores NKT-cell function in an in vitro ovarian cancer model. These studies suggest that the combination of immune modulation with antiangiogenic treatment has therapeutic potential in ovarian cancer. Clin Cancer Res; 22(16); 4249-58. ©2016 AACR.
Collapse
Affiliation(s)
- Irina V Tiper
- Department of Microbiology and Immunology, University of Maryland School of Medicine, Baltimore, Maryland
| | - Sarah M Temkin
- Department of Medicine, University of Maryland School of Medicine, Baltimore, Maryland. Department of Gynecology and Obstetrics, The Kelly Gynecologic Oncology Service, The Sidney Kimmel Comprehensive Cancer Center, The Johns Hopkins School of Medicine, Baltimore, Maryland
| | - Sarah Spiegel
- Department of Microbiology and Immunology, University of Maryland School of Medicine, Baltimore, Maryland
| | - Simeon E Goldblum
- Department of Medicine, University of Maryland School of Medicine, Baltimore, Maryland
| | - Robert L Giuntoli
- Department of Gynecology and Obstetrics, The Kelly Gynecologic Oncology Service, The Sidney Kimmel Comprehensive Cancer Center, The Johns Hopkins School of Medicine, Baltimore, Maryland
| | - Mathias Oelke
- Department of Pathology, The Johns Hopkins School of Medicine, Baltimore, Maryland
| | - Jonathan P Schneck
- Department of Pathology, The Johns Hopkins School of Medicine, Baltimore, Maryland
| | - Tonya J Webb
- Department of Microbiology and Immunology, University of Maryland School of Medicine, Baltimore, Maryland.
| |
Collapse
|
434
|
Guldner IH, Yang L, Cowdrick KR, Wang Q, Alvarez Barrios WV, Zellmer VR, Zhang Y, Host M, Liu F, Chen DZ, Zhang S. An Integrative Platform for Three-dimensional Quantitative Analysis of Spatially Heterogeneous Metastasis Landscapes. Sci Rep 2016; 6:24201. [PMID: 27068335 PMCID: PMC4828720 DOI: 10.1038/srep24201] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2015] [Accepted: 03/23/2016] [Indexed: 01/12/2023] Open
Abstract
Metastatic microenvironments are spatially and compositionally heterogeneous. This seemingly stochastic heterogeneity provides researchers great challenges in elucidating factors that determine metastatic outgrowth. Herein, we develop and implement an integrative platform that will enable researchers to obtain novel insights from intricate metastatic landscapes. Our two-segment platform begins with whole tissue clearing, staining, and imaging to globally delineate metastatic landscape heterogeneity with spatial and molecular resolution. The second segment of our platform applies our custom-developed SMART 3D (Spatial filtering-based background removal and Multi-chAnnel forest classifiers-based 3D ReconsTruction), a multi-faceted image analysis pipeline, permitting quantitative interrogation of functional implications of heterogeneous metastatic landscape constituents, from subcellular features to multicellular structures, within our large three-dimensional (3D) image datasets. Coupling whole tissue imaging of brain metastasis animal models with SMART 3D, we demonstrate the capability of our integrative pipeline to reveal and quantify volumetric and spatial aspects of brain metastasis landscapes, including diverse tumor morphology, heterogeneous proliferative indices, metastasis-associated astrogliosis, and vasculature spatial distribution. Collectively, our study demonstrates the utility of our novel integrative platform to reveal and quantify the global spatial and volumetric characteristics of the 3D metastatic landscape with unparalleled accuracy, opening new opportunities for unbiased investigation of novel biological phenomena in situ.
Collapse
Affiliation(s)
- Ian H Guldner
- Department of Biological Sciences, College of Science, University of Notre Dame, Notre Dame, IN 46556, USA.,Mike and Josie Harper Cancer Research Institute, University of Notre Dame, 1234 N. Notre Dame Avenue, South Bend, IN 46617, USA
| | - Lin Yang
- Department of Computer Science and Engineering, College of Engineering, University of Notre Dame, Notre Dame, IN 46556, USA
| | - Kyle R Cowdrick
- Department of Biological Sciences, College of Science, University of Notre Dame, Notre Dame, IN 46556, USA.,Mike and Josie Harper Cancer Research Institute, University of Notre Dame, 1234 N. Notre Dame Avenue, South Bend, IN 46617, USA
| | - Qingfei Wang
- Department of Biological Sciences, College of Science, University of Notre Dame, Notre Dame, IN 46556, USA.,Mike and Josie Harper Cancer Research Institute, University of Notre Dame, 1234 N. Notre Dame Avenue, South Bend, IN 46617, USA
| | - Wendy V Alvarez Barrios
- Department of Biological Sciences, College of Science, University of Notre Dame, Notre Dame, IN 46556, USA.,Mike and Josie Harper Cancer Research Institute, University of Notre Dame, 1234 N. Notre Dame Avenue, South Bend, IN 46617, USA
| | - Victoria R Zellmer
- Department of Biological Sciences, College of Science, University of Notre Dame, Notre Dame, IN 46556, USA.,Mike and Josie Harper Cancer Research Institute, University of Notre Dame, 1234 N. Notre Dame Avenue, South Bend, IN 46617, USA
| | - Yizhe Zhang
- Department of Computer Science and Engineering, College of Engineering, University of Notre Dame, Notre Dame, IN 46556, USA
| | - Misha Host
- Department of Biological Sciences, College of Science, University of Notre Dame, Notre Dame, IN 46556, USA.,Mike and Josie Harper Cancer Research Institute, University of Notre Dame, 1234 N. Notre Dame Avenue, South Bend, IN 46617, USA
| | - Fang Liu
- Mike and Josie Harper Cancer Research Institute, University of Notre Dame, 1234 N. Notre Dame Avenue, South Bend, IN 46617, USA.,Department of Applied and Computational Mathematics and Statistics, College of Science, University of Notre Dame, Notre Dame, IN 46556, USA
| | - Danny Z Chen
- Mike and Josie Harper Cancer Research Institute, University of Notre Dame, 1234 N. Notre Dame Avenue, South Bend, IN 46617, USA.,Department of Computer Science and Engineering, College of Engineering, University of Notre Dame, Notre Dame, IN 46556, USA
| | - Siyuan Zhang
- Department of Biological Sciences, College of Science, University of Notre Dame, Notre Dame, IN 46556, USA.,Mike and Josie Harper Cancer Research Institute, University of Notre Dame, 1234 N. Notre Dame Avenue, South Bend, IN 46617, USA
| |
Collapse
|
435
|
Pitt JM, Marabelle A, Eggermont A, Soria JC, Kroemer G, Zitvogel L. Targeting the tumor microenvironment: removing obstruction to anticancer immune responses and immunotherapy. Ann Oncol 2016; 27:1482-92. [PMID: 27069014 DOI: 10.1093/annonc/mdw168] [Citation(s) in RCA: 870] [Impact Index Per Article: 96.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2016] [Accepted: 04/06/2016] [Indexed: 12/23/2022] Open
Abstract
The tumor microenvironment (TME) is an integral part of cancer. Recognition of the essential nature of the TME in cancer evolution has led to a shift from a tumor cell-centered view of cancer development to the concept of a complex tumor ecosystem that supports tumor growth and metastatic dissemination. Accordingly, novel targets within the TME have been uncovered that can help direct and improve the actions of various cancer therapies, notably immunotherapies that work by potentiating host antitumor immune responses. Here, we review the composition of the TME, how this attenuates immunosurveillance, and discuss existing and potential strategies aimed at targeting cellular and molecular TME components.
Collapse
Affiliation(s)
- J M Pitt
- Institut de Cancérologie Gustave Roussy Cancer Campus (GRCC), Villejuif INSERM Unit U1015, Villejuif Faculté de Médecine, Université Paris Sud, Université Paris-Saclay, Le Kremlin Bicêtre Gustave Roussy Cancer Campus, Villejuif Cedex
| | - A Marabelle
- Institut de Cancérologie Gustave Roussy Cancer Campus (GRCC), Villejuif INSERM Unit U1015, Villejuif INSERM Unit U981, Villejuif
| | - A Eggermont
- Institut de Cancérologie Gustave Roussy Cancer Campus (GRCC), Villejuif
| | - J-C Soria
- Institut de Cancérologie Gustave Roussy Cancer Campus (GRCC), Villejuif Faculté de Médecine, Université Paris Sud, Université Paris-Saclay, Le Kremlin Bicêtre INSERM Unit U981, Villejuif Drug Development Department (DITEP), Villejuif
| | - G Kroemer
- INSERM U848, Villejuif Metabolomics Platform, GRCC, Villejuif Equipe 11 labellisée Ligue contre le Cancer, Centre de Recherche des Cordeliers, INSERM U 1138, Paris Pôle de Biologie, Hôpital Européen Georges Pompidou, AP-HP, Paris Université Paris Descartes, Sorbonne Paris Cité, Paris Université Pierre et Marie Curie, Paris, France Department of Women's and Children's Health, Karolinska Institute, Karolinska University Hospital, Stockholm, Sweden
| | - L Zitvogel
- Institut de Cancérologie Gustave Roussy Cancer Campus (GRCC), Villejuif INSERM Unit U1015, Villejuif Faculté de Médecine, Université Paris Sud, Université Paris-Saclay, Le Kremlin Bicêtre INSERM Unit U932, Institut Curie, Paris Cedex 05 Center of Clinical Investigations in Biotherapies of Cancer (CICBT) 507, Villejuif, France
| |
Collapse
|
436
|
Helal-Neto E, Brandão-Costa RM, Saldanha-Gama R, Ribeiro-Pereira C, Midlej V, Benchimol M, Morandi V, Barja-Fidalgo C. Priming Endothelial Cells With a Melanoma-Derived Extracellular Matrix Triggers the Activation of αvβ3/VEGFR2 Axis. J Cell Physiol 2016; 231:2464-73. [DOI: 10.1002/jcp.25358] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2015] [Accepted: 02/22/2016] [Indexed: 12/14/2022]
Affiliation(s)
- Edward Helal-Neto
- Laboratório de Farmacologia Celular e Molecular, IBRAG; Universidade do Estado do Rio de Janeiro; Rio de Janeiro Brazil
- Laboratório de Biologia da Célula Endotelial e da Angiogênese, IBRAG; Universidade do Estado do Rio de Janeiro; Rio de Janeiro Brazil
| | - Renata M. Brandão-Costa
- Laboratório de Farmacologia Celular e Molecular, IBRAG; Universidade do Estado do Rio de Janeiro; Rio de Janeiro Brazil
| | - Roberta Saldanha-Gama
- Laboratório de Farmacologia Celular e Molecular, IBRAG; Universidade do Estado do Rio de Janeiro; Rio de Janeiro Brazil
| | - Cristiane Ribeiro-Pereira
- Laboratório de Farmacologia Celular e Molecular, IBRAG; Universidade do Estado do Rio de Janeiro; Rio de Janeiro Brazil
| | - Victor Midlej
- Instituto de Biofísica Carlos Chagas Filho; Universidade Federal do Rio de Janeiro; Rio de Janeiro Brazil
| | - Marlene Benchimol
- Instituto de Biofísica Carlos Chagas Filho; Universidade Federal do Rio de Janeiro; Rio de Janeiro Brazil
- Unigranrio; Universidade do Grande Rio; Rio de Janeiro Brazil
| | - Verônica Morandi
- Laboratório de Biologia da Célula Endotelial e da Angiogênese, IBRAG; Universidade do Estado do Rio de Janeiro; Rio de Janeiro Brazil
| | - Christina Barja-Fidalgo
- Laboratório de Farmacologia Celular e Molecular, IBRAG; Universidade do Estado do Rio de Janeiro; Rio de Janeiro Brazil
| |
Collapse
|
437
|
Martial S. Involvement of ion channels and transporters in carcinoma angiogenesis and metastasis. Am J Physiol Cell Physiol 2016; 310:C710-27. [PMID: 26791487 DOI: 10.1152/ajpcell.00218.2015] [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] [Indexed: 12/21/2022]
Abstract
Angiogenesis is a finely tuned process, which is the result of the equilibrium between pro- and antiangiogenic factors. In solid tumor angiogenesis, the balance is highly in favor of the production of new, but poorly functional blood vessels, initially intended to provide growing tumors with nutrients and oxygen. Among the numerous proteins involved in tumor development, several types of ion channels are overexpressed in tumor cells, as well as in stromal and endothelial cells. Ion channels thus actively participate in the different hallmarks of cancer, especially in tumor angiogenesis and metastasis. Indeed, from their strategic localization in the plasma membrane, ion channels are key operators of cell signaling, as they sense and respond to environmental changes. This review aims to decipher how ion channels of different families are intricately involved in the fundamental angiogenesis and metastasis hallmarks, which lead from a nascent tumor to systemic dissemination. An overview of the possible use of ion channels as therapeutic targets will also be given, showing that ion channel inhibitors or specific antibodies may provide effective tools, in the near future, in the treatment of carcinomas.
Collapse
Affiliation(s)
- Sonia Martial
- Institut de Recherche sur le Cancer et le Vieillissement, CNRS UMR 7284, Inserm U1081, Université Nice-Sophia Antipolis, Nice, France
| |
Collapse
|
438
|
Potential biomarker for checkpoint blockade immunotherapy and treatment strategy. Tumour Biol 2016; 37:4251-61. [PMID: 26779629 DOI: 10.1007/s13277-016-4812-9] [Citation(s) in RCA: 66] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2015] [Accepted: 01/06/2016] [Indexed: 12/12/2022] Open
Abstract
Programmed cell death protein-1 (PD-1) and ligand (PD-L1) provide an important escape mechanism from immune attack, and blockade therapy of these proteins show promising clinical benefits in many types of cancer. PD-L1 can be induced by interferon-gamma (IFN-γ), hypoxia, or toll-like receptor (TLR)-mediated pathways that confer adaptive immune resistance, or upregulated by oncogenic signals leading to constitutive expression and resulting in intrinsic immune resistance. The PD-1/PD-L1 checkpoint blockade, which targets regulatory pathways in T cells to overcome immune resistance, is correlated to PD-L1 expression pattern and the presence of tumor-infiltrating lymphocytes (TILs). Meanwhile, immunogenic mutation loads show significant response to checkpoint blockade, which is probably due to PD-1/L1 status and TIL content. Finally, the clinical strategies to design effective checkpoint-targeting immunotherapies are based on the classification of inducible/constitutive expression of PD-L1 and the presence of TILs.
Collapse
|
439
|
Eccles SA, Court W, Patterson L. In Vitro Assays for Endothelial Cell Functions Required for Angiogenesis: Proliferation, Motility, Tubular Differentiation, and Matrix Proteolysis. Methods Mol Biol 2016; 1430:121-147. [PMID: 27172950 DOI: 10.1007/978-1-4939-3628-1_8] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/05/2023]
Abstract
This chapter deconstructs the process of angiogenesis into its component parts in order to provide simple assays to measure discrete endothelial cell functions. The techniques described will be suitable for studying stimulators and/or inhibitors of angiogenesis and determining which aspect of the process is modulated. The assays are designed to be robust and straightforward, using human umbilical vein endothelial cells, but with an option to use other sources such as microvascular endothelial cells from various tissues or lymphatic endothelial cells. It must be appreciated that such reductionist approaches cannot cover the complexity of the angiogenic process as a whole, incorporating as it does a myriad of positive and negative signals, three-dimensional interactions with host tissues and many accessory cells including fibroblasts, macrophages, pericytes and platelets. The extent to which in vitro assays predict physiological or pathological processes in vivo (e.g., wound healing, tumor angiogenesis) or surrogate techniques such as the use of Matrigel™ plugs, sponge implants, corneal assays etc remains to be determined.
Collapse
Affiliation(s)
- Suzanne A Eccles
- Cancer Research UK Cancer Therapeutics Unit, Centre for Cancer Imaging, The Institute of Cancer Research, Cotswold Rd., Sutton, Surrey, SM2 5NG, UK.
| | - William Court
- Cancer Research UK Cancer Therapeutics Unit, Centre for Cancer Imaging, The Institute of Cancer Research, Cotswold Rd., Sutton, Surrey, SM2 5NG, UK
| | - Lisa Patterson
- Cancer Research UK Cancer Therapeutics Unit, Centre for Cancer Imaging, The Institute of Cancer Research, Cotswold Rd., Sutton, Surrey, SM2 5NG, UK
| |
Collapse
|
440
|
Kouhsoltani M, Moradzadeh Khiavi M, Jamali G, Farnia S. Immunohistochemical Assessment of Mast Cells and Small Blood Vessels in Dentigerous Cyst, Odontogenic Keratocyst, and Periapical Cyst. Adv Pharm Bull 2015; 5:637-41. [PMID: 26793609 DOI: 10.15171/apb.2015.086] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2015] [Revised: 08/31/2015] [Accepted: 09/07/2015] [Indexed: 11/09/2022] Open
Abstract
PURPOSE The aim of this study was to verify the density of mast cells (MCs) and microvessels in odontogenic cysts. Furthermore, the correlation between MCs and microvessels was evaluated to assess the contribution of MCs to angiogenesis and growth of odontogenic cysts. This approach may be a basis for the development of future pharmaceuticals addressed to MCs performance to manage odontogenic cysts. To our knowledge, no study investigating the correlation between MCs and microvessels has been performed to date. METHODS 60 cases of odontogenic cysts consisting of 20 radicular cysts (RCs), 20 odontogenic keratocysts (OKCs) and 20 dentigerous cysts (DCs) were included in this study. Five high power fields in superficial connective tissue and five high power fields in deep connective tissue were counted for each sample. Moreover, a total mean of ten fields was calculated. RESULTS RC showed the highest mean numbers of MCs and microvessels (p<0.05). The subepithelial zones of all cysts contained more MCs and microvessels compared to the deeper zones. A statistically significant correlation between the numbers of MCs and microvessels was not observed (r=0.00, p=0.49). CONCLUSION Although the number of MCs was not significantly associated with microvessels, these cells may be related to the growth of odontogenic lesions, particularly RCs. Further studies on the in vivo functions of MCs will make the concept more clear.
Collapse
Affiliation(s)
- Maryam Kouhsoltani
- Dental and Periodontal Research Center and Department of Oral and Maxillofacial Pathology, Faculty of Dentistry, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Monir Moradzadeh Khiavi
- Department of Oral and Maxillofacial Pathology, Dental Faculty, Tehran University of Medical Sciences, Tehran, Iran
| | - Golshan Jamali
- Department of Radiology, Dental Faculty, Shahid Sadoughi University of Medical Sciences, Yazd, Iran
| | - Samira Farnia
- Drug Applied Research Center and Department of Oral and Maxillofacial Pathology, Faculty of Dentistry, Tabriz University of Medical Sciences, Tabriz, Iran
| |
Collapse
|
441
|
Maes H, Kuchnio A, Carmeliet P, Agostinis P. Chloroquine anticancer activity is mediated by autophagy-independent effects on the tumor vasculature. Mol Cell Oncol 2015; 3:e970097. [PMID: 27308577 PMCID: PMC4845249 DOI: 10.4161/23723548.2014.970097] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2014] [Revised: 09/10/2014] [Accepted: 09/10/2014] [Indexed: 11/19/2022]
Abstract
Chloroquine is used clinically as an autophagy blocker to potentiate anticancer treatments. However, whether chloroquine acts solely through autophagy-dependent and cancer cell autonomous mechanisms has remained elusive. In a recent study we found that chloroquine reduced intratumoral hypoxia and metastasis, while improving chemotherapy response, largely through an autophagy-independent, NOTCH1-reliant mechanism of tumor vessel normalization.
Collapse
Affiliation(s)
- Hannelore Maes
- Department Cellular and Molecular Medicine; Laboratory of Cell Death and Therapy ; KU Leuven, Leuven, Belgium
| | - Anna Kuchnio
- Department of Oncology; Laboratory of Angiogenesis and Neurovascular Link; KU Leuven, Leuven, Belgium; Vesalius Research Center; Laboratory of Angiogenesis and Neurovascular Link; VIB, Leuven, Belgium
| | - Peter Carmeliet
- Department of Oncology; Laboratory of Angiogenesis and Neurovascular Link; KU Leuven, Leuven, Belgium; Vesalius Research Center; Laboratory of Angiogenesis and Neurovascular Link; VIB, Leuven, Belgium
| | - Patrizia Agostinis
- Department Cellular and Molecular Medicine; Laboratory of Cell Death and Therapy ; KU Leuven, Leuven, Belgium
| |
Collapse
|
442
|
|
443
|
Johansson-Percival A, Li ZJ, Lakhiani D, He B, Wang X, Hamzah J, Ganss R. Intratumoral LIGHT Restores Pericyte Contractile Properties and Vessel Integrity. Cell Rep 2015; 13:2687-98. [DOI: 10.1016/j.celrep.2015.12.004] [Citation(s) in RCA: 55] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2015] [Revised: 10/12/2015] [Accepted: 11/19/2015] [Indexed: 12/11/2022] Open
|
444
|
Pikor LA, Bell JC, Diallo JS. Oncolytic Viruses: Exploiting Cancer's Deal with the Devil. Trends Cancer 2015; 1:266-277. [PMID: 28741515 DOI: 10.1016/j.trecan.2015.10.004] [Citation(s) in RCA: 70] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2015] [Revised: 10/18/2015] [Accepted: 10/20/2015] [Indexed: 12/12/2022]
Abstract
Tumor cells harbor tens to thousands of genetic and epigenetic alterations that disrupt cellular pathways, providing them with growth and survival advantages. However, these benefits come at a cost, with uncontrolled cell growth, defective apoptosis, sustained pathological angiogenesis, immune evasion, and a metastatic phenotype occurring at the expense of the antiviral response of the individual tumor cell. Oncolytic virotherapy is an emerging therapeutic strategy that uses replication-competent viruses to selectivity kill cancer cells by exploiting their impaired antiviral response. In this review, we outline our understanding of the alterations in signaling pathways that simultaneously contribute to the malignant phenotype and virus-mediated killing of cancer cells.
Collapse
Affiliation(s)
- Larissa A Pikor
- Center for Innovative Cancer Therapeutics, Ottawa Hospital Research Institute, Ottawa, ONT, Canada
| | - John C Bell
- Center for Innovative Cancer Therapeutics, Ottawa Hospital Research Institute, Ottawa, ONT, Canada; Department of Biochemistry, Immunology and Microbiology, University of Ottawa, Ottawa, ONT, Canada
| | - Jean-Simon Diallo
- Center for Innovative Cancer Therapeutics, Ottawa Hospital Research Institute, Ottawa, ONT, Canada.
| |
Collapse
|
445
|
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: 1.6] [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
|
446
|
Maes H, Olmeda D, Soengas MS, Agostinis P. Vesicular trafficking mechanisms in endothelial cells as modulators of the tumor vasculature and targets of antiangiogenic therapies. FEBS J 2015; 283:25-38. [PMID: 26443003 DOI: 10.1111/febs.13545] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2015] [Revised: 09/21/2015] [Accepted: 10/02/2015] [Indexed: 11/25/2022]
Abstract
A common feature of solid tumors is their ability to incite the formation of new blood and lymph vessels trough the processes of angiogenesis and lymphangiogenesis, respectively, to support tumor growth and favor metastatic dissemination. As a result of the lack of feedback regulatory control mechanisms or due to the exacerbated presence of pro-angiogenic signals within the tumor microenvironment, the tumor endothelium receives continuous signals to sprout and develop, generating vessels that are structurally and functionally abnormal. An emerging mechanism playing a central role in shaping the tumor vasculature is the endothelial-vesicular network that regulates trafficking/export and degradation of key signaling proteins and membrane receptors, including the vascular endothelial growth-factor receptor-2/3 and members of the Notch pathway. Here we will discuss recent evidence highlighting how vesicular trafficking mechanisms in endothelial cells contribute to pathological angiogenesis/lymphangiogenesis and can provide novel and exploitable targets in antiangiogenic therapies.
Collapse
Affiliation(s)
- Hannelore Maes
- Cell Death Research & Therapy (CDRT) Unit, Department of Cellular and Molecular Medicine, KU Leuven University of Leuven, Belgium
| | - David Olmeda
- Melanoma Laboratory, Molecular Oncology Programme, Spanish National Cancer Research Centre (CNIO), Madrid, Spain
| | - María S Soengas
- Melanoma Laboratory, Molecular Oncology Programme, Spanish National Cancer Research Centre (CNIO), Madrid, Spain
| | - Patrizia Agostinis
- Cell Death Research & Therapy (CDRT) Unit, Department of Cellular and Molecular Medicine, KU Leuven University of Leuven, Belgium
| |
Collapse
|
447
|
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
|
448
|
Gkretsi V, Stylianou A, Papageorgis P, Polydorou C, Stylianopoulos T. Remodeling Components of the Tumor Microenvironment to Enhance Cancer Therapy. Front Oncol 2015; 5:214. [PMID: 26528429 PMCID: PMC4604307 DOI: 10.3389/fonc.2015.00214] [Citation(s) in RCA: 93] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2015] [Accepted: 09/22/2015] [Indexed: 12/18/2022] Open
Abstract
Solid tumor pathophysiology is characterized by an abnormal microenvironment that guides tumor progression and poses barriers to the efficacy of cancer therapies. Most common among tumor types are abnormalities in the structure of the tumor vasculature and stroma. Remodeling the tumor microenvironment with the aim to normalize any aberrant properties has the potential to improve therapy. In this review, we discuss structural abnormalities of the tumor microenvironment and summarize the therapeutic strategies that have been developed to normalize tumors as well as their potential to enhance therapy. Finally, we present different in vitro models that have been developed to analyze and better understand the effects of the tumor microenvironment on cancer cell behavior.
Collapse
Affiliation(s)
- Vasiliki Gkretsi
- Cancer Biophysics Laboratory, Department of Mechanical and Manufacturing Engineering, University of Cyprus , Nicosia , Cyprus
| | - Andreas Stylianou
- Cancer Biophysics Laboratory, Department of Mechanical and Manufacturing Engineering, University of Cyprus , Nicosia , Cyprus
| | - Panagiotis Papageorgis
- Cancer Biophysics Laboratory, Department of Mechanical and Manufacturing Engineering, University of Cyprus , Nicosia , Cyprus ; Program in Biological Sciences, Department of Health Sciences, European University Cyprus , Nicosia , Cyprus
| | - Christiana Polydorou
- Cancer Biophysics Laboratory, Department of Mechanical and Manufacturing Engineering, University of Cyprus , Nicosia , Cyprus
| | - Triantafyllos Stylianopoulos
- Cancer Biophysics Laboratory, Department of Mechanical and Manufacturing Engineering, University of Cyprus , Nicosia , Cyprus
| |
Collapse
|
449
|
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
|
450
|
Gao DY, Lin TT, Sung YC, Liu YC, Chiang WH, Chang CC, Liu JY, Chen Y. CXCR4-targeted lipid-coated PLGA nanoparticles deliver sorafenib and overcome acquired drug resistance in liver cancer. Biomaterials 2015. [DOI: 10.1016/j.biomaterials.2015.07.035] [Citation(s) in RCA: 84] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
|