101
|
Van der Jeught K, Bialkowski L, Daszkiewicz L, Broos K, Goyvaerts C, Renmans D, Van Lint S, Heirman C, Thielemans K, Breckpot K. Targeting the tumor microenvironment to enhance antitumor immune responses. Oncotarget 2015; 6:1359-81. [PMID: 25682197 PMCID: PMC4359300 DOI: 10.18632/oncotarget.3204] [Citation(s) in RCA: 52] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2014] [Accepted: 12/24/2014] [Indexed: 12/16/2022] Open
Abstract
The identification of tumor-specific antigens and the immune responses directed against them has instigated the development of therapies to enhance antitumor immune responses. Most of these cancer immunotherapies are administered systemically rather than directly to tumors. Nonetheless, numerous studies have demonstrated that intratumoral therapy is an attractive approach, both for immunization and immunomodulation purposes. Injection, recruitment and/or activation of antigen-presenting cells in the tumor nest have been extensively studied as strategies to cross-prime immune responses. Moreover, delivery of stimulatory cytokines, blockade of inhibitory cytokines and immune checkpoint blockade have been explored to restore immunological fitness at the tumor site. These tumor-targeted therapies have the potential to induce systemic immunity without the toxicity that is often associated with systemic treatments. We review the most promising intratumoral immunotherapies, how these affect systemic antitumor immunity such that disseminated tumor cells are eliminated, and which approaches have been proven successful in animal models and patients.
Collapse
Affiliation(s)
- Kevin Van der Jeught
- Laboratory of Molecular and Cellular Therapy, Department of Immunology-Physiology, Vrije Universiteit Brussel, Laarbeeklaan, Jette, Belgium
| | - Lukasz Bialkowski
- Laboratory of Molecular and Cellular Therapy, Department of Immunology-Physiology, Vrije Universiteit Brussel, Laarbeeklaan, Jette, Belgium
| | - Lidia Daszkiewicz
- Laboratory of Molecular and Cellular Therapy, Department of Immunology-Physiology, Vrije Universiteit Brussel, Laarbeeklaan, Jette, Belgium
| | - Katrijn Broos
- Laboratory of Molecular and Cellular Therapy, Department of Immunology-Physiology, Vrije Universiteit Brussel, Laarbeeklaan, Jette, Belgium
| | - Cleo Goyvaerts
- Laboratory of Molecular and Cellular Therapy, Department of Immunology-Physiology, Vrije Universiteit Brussel, Laarbeeklaan, Jette, Belgium
| | - Dries Renmans
- Laboratory of Molecular and Cellular Therapy, Department of Immunology-Physiology, Vrije Universiteit Brussel, Laarbeeklaan, Jette, Belgium
| | - Sandra Van Lint
- Laboratory of Molecular and Cellular Therapy, Department of Immunology-Physiology, Vrije Universiteit Brussel, Laarbeeklaan, Jette, Belgium
| | - Carlo Heirman
- Laboratory of Molecular and Cellular Therapy, Department of Immunology-Physiology, Vrije Universiteit Brussel, Laarbeeklaan, Jette, Belgium
| | - Kris Thielemans
- Laboratory of Molecular and Cellular Therapy, Department of Immunology-Physiology, Vrije Universiteit Brussel, Laarbeeklaan, Jette, Belgium
| | - Karine Breckpot
- Laboratory of Molecular and Cellular Therapy, Department of Immunology-Physiology, Vrije Universiteit Brussel, Laarbeeklaan, Jette, Belgium
| |
Collapse
|
102
|
Kobayashi T, Doff BL, Rearden RC, Leggatt GR, Mattarollo SR. NKT cell-targeted vaccination plus anti-4-1BB antibody generates persistent CD8 T cell immunity against B cell lymphoma. Oncoimmunology 2015; 4:e990793. [PMID: 25949907 PMCID: PMC4404843 DOI: 10.4161/2162402x.2014.990793] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2014] [Accepted: 11/18/2014] [Indexed: 12/28/2022] Open
Abstract
Harnessing the immune adjuvant properties of natural killer T (NKT) cells is an effective strategy to generate anticancer immunity. The objective of this study was to increase the potency and durability of vaccine-induced immunity against B cell lymphoma by combining α-galactosylceramide (α-GalCer)-loaded tumor cell vaccination with an agonistic antibody targeting the immune checkpoint molecule 4–1BB (CD137). We observed potent synergy when combining vaccination and anti-4–1BB antibody treatment resulting in significantly enhanced survival of mice harboring Eμ-myc tumors, including complete eradication of lymphoma in over 50% of mice. Tumor-free survival required interferon γ (IFNγ)-dependent expansion of CD8+ T cells and was associated with 4–1BB-mediated differentiation of KLRG1+ effector CD8+ T cells. 'Cured' mice were also resistant to lymphoma re-challenge 80 days later indicating successful generation of immunological memory. Overall, our results demonstrate that therapeutic anticancer vaccination against B cell lymphoma using an NKT cell ligand can be boosted by subsequent co-stimulation through 4–1BB leading to a sustainable immune response that may enhance outcomes to conventional treatment.
Collapse
Affiliation(s)
- Takumi Kobayashi
- The University of Queensland Diamantina Institute; The University of Queensland; Translational Research Institute, Brisbane ; Queensland, Australia
| | - Brianna L Doff
- The University of Queensland Diamantina Institute; The University of Queensland; Translational Research Institute, Brisbane ; Queensland, Australia
| | - Rory C Rearden
- The University of Queensland Diamantina Institute; The University of Queensland; Translational Research Institute, Brisbane ; Queensland, Australia
| | - Graham R Leggatt
- The University of Queensland Diamantina Institute; The University of Queensland; Translational Research Institute, Brisbane ; Queensland, Australia
| | - Stephen R Mattarollo
- The University of Queensland Diamantina Institute; The University of Queensland; Translational Research Institute, Brisbane ; Queensland, Australia
| |
Collapse
|
103
|
Chester C, Marabelle A, Houot R, Kohrt HE. Dual antibody therapy to harness the innate anti-tumor immune response to enhance antibody targeting of tumors. Curr Opin Immunol 2015; 33:1-8. [PMID: 25576932 DOI: 10.1016/j.coi.2014.12.010] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2014] [Revised: 12/19/2014] [Accepted: 12/24/2014] [Indexed: 01/05/2023]
Abstract
Cancer immunotherapy is a rapidly evolving field that offers a novel paradigm for cancer treatment: therapies focus on enhancing the immune system's innate and adaptive anti-tumor response. Early immunotherapeutics have achieved impressive clinical outcomes and monoclonal antibodies are now integral to therapeutic strategies in a variety of cancers. However, only recently have antibodies targeting innate immune cells entered clinical development. Innate immune effector cells play important roles in generating and maintaining antitumor immunity. Antibody-dependent cell-mediated cytotoxicity (ADCC) and antibody-dependent cellular phagocytosis (ADCP) are important innate immune mechanisms for tumor eradication. These cytolytic processes are initiated by the detection of a tumor-targeting antibody and can be augmented by activating co-stimulatory pathways or blocking inhibitory signals on innate immune cells. The combination of FDA-approved monoclonal antibodies with innate effector-targeting antibodies has demonstrated potent preclinical therapeutic synergy and early-phase combinatorial clinical trials are ongoing.
Collapse
Affiliation(s)
- Cariad Chester
- Department of Medicine, Division of Oncology, Stanford University, Stanford, CA 94305, USA
| | | | - Roch Houot
- Service d'Hématologie Clinique, Centre Hospitalier Universitaire de Rennes, Rennes, France; CHU Rennes, Service Hématologie Clinique, F-35033 Rennes, France; INSERM, U917, F-35043 Rennes, France
| | - Holbrook E Kohrt
- Department of Medicine, Division of Oncology, Stanford University, Stanford, CA 94305, USA.
| |
Collapse
|
104
|
Massari F, Santoni M, Ciccarese C, Santini D, Alfieri S, Martignoni G, Brunelli M, Piva F, Berardi R, Montironi R, Porta C, Cascinu S, Tortora G. PD-1 blockade therapy in renal cell carcinoma: current studies and future promises. Cancer Treat Rev 2015; 41:114-21. [PMID: 25586601 DOI: 10.1016/j.ctrv.2014.12.013] [Citation(s) in RCA: 139] [Impact Index Per Article: 15.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2014] [Accepted: 12/30/2014] [Indexed: 10/24/2022]
Abstract
RCC is considered an immunogenic tumor with a prominent dysfunctional immune cell infiltrate, unable to control tumor growth. Evasion of immune surveillance, a process defined immune-editing, leads to malignant progression. The striking improvement of knowledge in immunology has led to the identification of immune checkpoints (such as CTLA-4 and PD-1), whose blockage enhances the antitumor immunity. The interaction between PD-1, an inducible inhibitory receptor expressed on lymphocytes and DCs, and PD-L1 ligand, expressed by tumor cells, results in a down-regulation of the T-cell response. Therefore, the PD-1/PD-L1 axis inhibition by targeted-antibodies, increasing the T-cell proliferation and cytotoxicity, represents a promising mechanism to stimulate the anti-tumor activity of the immune system, improving the outcomes of cancer patients. Several PD-1 and PD-L1 inhibitors have been evaluated in different tumor types, showing promising results. The interesting correlation between lymphocytes PD-1 expression and RCC advanced stage, grade and prognosis, as well as the selective PD-L1 expression by RCC tumor cells and its potential association with worse clinical outcomes, have led to the development of new anti PD-1/PD-L1 agents, alone or in combination with anti-angiogenic drugs or other immunotherapeutic approaches, for the treatment of RCC. In this review we discuss the role of PD-1/PD-L1 in RCC, focusing on the biological rationale, current clinical studies and promising therapeutic perspectives to target the PD-1 pathway.
Collapse
Affiliation(s)
- F Massari
- Medical Oncology, Azienda Ospedaliera Universitaria Integrata, University of Verona, Piazzale L.A. Scuro 10, 37124 Verona, Italy
| | - M Santoni
- Clinica di Oncologia Medica, AOU "Ospedali Riuniti", Polytechnic University of the Marche Region, via Conca 71, 60126 Ancona, Italy.
| | - C Ciccarese
- Medical Oncology, Azienda Ospedaliera Universitaria Integrata, University of Verona, Piazzale L.A. Scuro 10, 37124 Verona, Italy
| | - D Santini
- Department of Medical Oncology, University Campus Bio-Medico Roma, Oncologia Medica, Rome, Italy
| | - S Alfieri
- SSD Oncologia medica Tumori Testa e Collo, Fondazione IRCCS - Istituto Nazionale dei Tumori, Via Venezian, 1, 20133 Milano, Italy
| | - G Martignoni
- Department of Pathology and Diagnostic, Azienda Ospedaliera Universitaria Integrata, University of Verona, Piazzale L.A. Scuro 10, 37124 Verona, Italy
| | - M Brunelli
- Department of Pathology and Diagnostic, Azienda Ospedaliera Universitaria Integrata, University of Verona, Piazzale L.A. Scuro 10, 37124 Verona, Italy
| | - F Piva
- Department of Specialistic Clinical and Odontostomatological Sciences, Polytechnic University of Marche, 60131 Ancona, Italy
| | - R Berardi
- Clinica di Oncologia Medica, AOU "Ospedali Riuniti", Polytechnic University of the Marche Region, via Conca 71, 60126 Ancona, Italy
| | - R Montironi
- Section of Pathological Anatomy, Polytechnic University of the Marche Region, School of Medicine, United Hospitals, Ancona, Italy
| | - C Porta
- Medical Oncology, IRCCS San Matteo University Hospital Foundation, piazzale C. Golgi 19, 27100 Pavia, Italy
| | - S Cascinu
- Clinica di Oncologia Medica, AOU "Ospedali Riuniti", Polytechnic University of the Marche Region, via Conca 71, 60126 Ancona, Italy
| | - G Tortora
- Medical Oncology, Azienda Ospedaliera Universitaria Integrata, University of Verona, Piazzale L.A. Scuro 10, 37124 Verona, Italy
| |
Collapse
|
105
|
Vinay DS, Kwon BS. 4-1BB (CD137), an inducible costimulatory receptor, as a specific target for cancer therapy. BMB Rep 2014; 47:122-9. [PMID: 24499671 PMCID: PMC4163883 DOI: 10.5483/bmbrep.2014.47.3.283] [Citation(s) in RCA: 86] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2013] [Revised: 12/30/2013] [Accepted: 01/28/2014] [Indexed: 01/09/2023] Open
Abstract
Although considerable progress has been made in understanding how tumors evade immune surveillance, measures to counter the same have not kept pace with the advances made in designing effective strategies. 4-1BB (CD137; TNFRS9), an activation-induced costimulatory molecule, is an important regulator of immune responses. Targeting 4-1BB or its natural ligand 4-1BB ligand (4-1BBL) has important implications in many clinical conditions, including cancer. In-depth analysis revealed that 4-1BB-mediated anti-cancer effects are based on its ability to induce activation of cytotoxic T lymphocytes (CTL), and among others, high amounts of IFN-γ. In this review, we will discuss the various aspects of 4-1BB-mediated anti-tumor responses, the basis of such responses, and future directions. [BMB Reports 2014; 47(3): 122-129]
Collapse
Affiliation(s)
- Dass S Vinay
- Section of Clinical Immunology, Allergy, and Rheumatology, Department of Medicine, Tulane University Health Sciences Center, New Orleans, LA70112, USA
| | - Byoung S Kwon
- Section of Clinical Immunology, Allergy, and Rheumatology, Department of Medicine, Tulane University Health Sciences Center, New Orleans, LA70112, USA; Cell and Immunobiology, and R & D Center for Cancer Therapeutics, National Cancer Center, Goyang 410-769, Korea
| |
Collapse
|
106
|
Abstract
In addition to T-cell receptor signals, T lymphocytes require costimulatory signals for robust activation. Among these, those mediated by 4-1BB (CD137, TNFRSF9) are critical for tumor immunity. 4-1BB is expressed in T-cell receptor-activated lymphocytes as well as natural killer cells and other hematopoietic and nonhematopoietic cells. 4-1BB ligation induces a signaling cascade that results in cytokine production, expression of antiapoptotic molecules, and enhanced immune responses. In line with the described function of 4-1BB, its addition to CD3ζ chimeric antigen receptors (CARs) increases their capacity to provoke T-cell expansion and antitumor activity. The results of preclinical studies with 4-1BB CARs have been corroborated by encouraging results from clinical trials. Advantages and disadvantages of 4-1BB CARs versus CARs bearing other costimulatory components remain to be fully elucidated. In this review, we discuss the properties of 4-1BB, the design of 4-1BB CARs, and the function of T lymphocytes and natural killer cells expressing them.
Collapse
|
107
|
Chen S, Lee LF, Fisher TS, Jessen B, Elliott M, Evering W, Logronio K, Tu GH, Tsaparikos K, Li X, Wang H, Ying C, Xiong M, VanArsdale T, Lin JC. Combination of 4-1BB agonist and PD-1 antagonist promotes antitumor effector/memory CD8 T cells in a poorly immunogenic tumor model. Cancer Immunol Res 2014; 3:149-60. [PMID: 25387892 DOI: 10.1158/2326-6066.cir-14-0118] [Citation(s) in RCA: 207] [Impact Index Per Article: 20.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Immunotherapies targeting the programmed death 1 (PD-1) coinhibitory receptor have shown great promise for a subset of patients with cancer. However, robust and safe combination therapies are still needed to bring the benefit of cancer immunotherapy to broader patient populations. To search for an optimal strategy of combinatorial immunotherapy, we have compared the antitumor activity of the anti-4-1BB/anti-PD-1 combination with that of the anti-PD-1/anti-LAG-3 combination in the poorly immunogenic B16F10 melanoma model. Pronounced tumor inhibition occurred only in animals receiving anti-PD-1 and anti-4-1BB concomitantly, while combining anti-PD-1 with anti-LAG-3 led to a modest degree of tumor suppression. The activity of the anti-4-1BB/anti-PD-1 combination was dependent on IFNγ and CD8(+) T cells. Both 4-1BB and PD-1 proteins were elevated on the surface of CD8(+) T cells by anti-4-1BB/anti-PD-1 cotreatment. In the tumor microenvironment, an effective antitumor immune response was induced as indicated by the increased CD8(+)/Treg ratio and the enrichment of genes such as Cd3e, Cd8a, Ifng, and Eomes. In the spleen, the combination treatment shaped the immune system to an effector/memory phenotype and increased the overall activity of tumor-specific CD8(+) CTLs, reflecting a long-lasting systemic antitumor response. Furthermore, combination treatment in C57BL/6 mice showed no additional safety signals, and only minimally increased severity of the known toxicity relative to 4-1BB agonist alone. Therefore, in the absence of any cancer vaccine, anti-4-1BB/anti-PD-1 combination therapy is sufficient to elicit a robust antitumor effector/memory T-cell response in an aggressive tumor model and is therefore a candidate for combination trials in patients.
Collapse
Affiliation(s)
- Shihao Chen
- Rinat Laboratories, Pfizer Inc., South San Francisco, California.
| | - Li-Fen Lee
- Rinat Laboratories, Pfizer Inc., South San Francisco, California
| | | | - Bart Jessen
- Drug Safety R&D, Pfizer Inc., San Diego, California
| | - Mark Elliott
- Oncology Research Unit, Pfizer Inc., San Diego, California
| | | | - Kathryn Logronio
- Rinat Laboratories, Pfizer Inc., South San Francisco, California
| | - Guang Huan Tu
- Rinat Laboratories, Pfizer Inc., South San Francisco, California
| | | | - Xiaoai Li
- Rinat Laboratories, Pfizer Inc., South San Francisco, California
| | - Hui Wang
- Oncology Research Unit, Pfizer Inc., San Diego, California
| | - Chi Ying
- Rinat Laboratories, Pfizer Inc., South San Francisco, California
| | - Mengli Xiong
- Rinat Laboratories, Pfizer Inc., South San Francisco, California
| | | | - John C Lin
- Rinat Laboratories, Pfizer Inc., South San Francisco, California.
| |
Collapse
|
108
|
Microenvironment of tumor-draining lymph nodes: opportunities for liposome-based targeted therapy. Int J Mol Sci 2014; 15:20209-39. [PMID: 25380524 PMCID: PMC4264163 DOI: 10.3390/ijms151120209] [Citation(s) in RCA: 48] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2014] [Revised: 10/20/2014] [Accepted: 10/24/2014] [Indexed: 02/07/2023] Open
Abstract
The World Health Organization (WHO) recently reported that the total number of global cancer cases in 2013 reached 14 million, a 10% rise since 2008, while the total number of cancer deaths reached 8.2 million, a 5.2% increase since 2008. Metastasis is the major cause of death from cancer, accounting for 90% of all cancer related deaths. Tumor-draining lymph nodes (TDLN), the sentinel nodes, are the first organs of metastasis in several types of cancers. The extent of metastasis in the TDLN is often used in disease staging and prognosis evaluation in cancer patients. Here, we describe the microenvironment of the TDLN and review the recent literature on liposome-based therapies directed to immune cells within the TDLN with the intent to target cancer cells.
Collapse
|
109
|
Park JW, Park DM, Choi BK, Kwon BS, Seong JK, Green JE, Kim DY, Kim HK. Establishment and characterization of metastatic gastric cancer cell lines from murine gastric adenocarcinoma lacking Smad4, p53, and E-cadherin. Mol Carcinog 2014; 54:1521-7. [DOI: 10.1002/mc.22226] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2014] [Revised: 08/06/2014] [Accepted: 08/13/2014] [Indexed: 02/04/2023]
Affiliation(s)
- Jun Won Park
- National Cancer Center; Goyang Gyeonggi Republic of Korea
- College of Veterinary Medicine; Seoul National University; Seoul Republic of Korea
| | - Dong Min Park
- National Cancer Center; Goyang Gyeonggi Republic of Korea
| | - Beom K. Choi
- National Cancer Center; Goyang Gyeonggi Republic of Korea
| | - Byoung S. Kwon
- National Cancer Center; Goyang Gyeonggi Republic of Korea
| | - Je Kyung Seong
- College of Veterinary Medicine; Seoul National University; Seoul Republic of Korea
| | - Jeffrey E. Green
- Laboratory of Cancer Biology and Genetics; National Cancer Institute; Bethesda Maryland
| | - Dae-Yong Kim
- College of Veterinary Medicine; Seoul National University; Seoul Republic of Korea
| | - Hark Kyun Kim
- National Cancer Center; Goyang Gyeonggi Republic of Korea
| |
Collapse
|
110
|
Demaria S, Pilones KA, Vanpouille-Box C, Golden EB, Formenti SC. The optimal partnership of radiation and immunotherapy: from preclinical studies to clinical translation. Radiat Res 2014; 182:170-81. [PMID: 24937779 PMCID: PMC4184032 DOI: 10.1667/rr13500.1] [Citation(s) in RCA: 69] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
The main role of the immune system is to restore tissue homeostasis when altered by pathogenic processes, including neoplastic transformation. Immune-mediated tumor rejection has been recognized as an extrinsic tumor suppressor mechanism that tumors need to overcome to progress. By the time a tumor becomes clinically apparent it has successfully escaped immune control by establishing an immunosuppressive microenvironment. Ionizing radiation applied locally to a tumor alters these tumor-host interactions. Accumulating evidence indicates that standard therapeutic doses of radiation have the potential to recover tumor immunogenicity and convert the tumor into an in situ personalized vaccine. Radiotherapy induces an immunogenic tumor cell death promoting cross-presentation of tumor-derived antigens by dendritic cells to T cells. In addition, radiotherapy stimulates chemokine-mediated recruitment of effector T cells to the tumor, and cellular recognition and killing by T cells that is facilitated by upregulation of major histocompatibility antigens, NKG2D ligands, adhesion molecules and death receptors. Despite these effects, radiotherapy alone is only rarely capable of generating enough proinflammatory signals to sufficiently overcome suppression, as it can also activate immunosuppressive factors. However, our group and others have shown that when combined with targeted immunotherapy agents radiotherapy significantly contributes to a therapeutically effective anti-tumor immune response. To illustrate this partnership between radiation and immunotherapy we will discuss as an example our experience in preclinical models and the molecular mechanisms identified. Additionally, the clinical translation of these combinations will be discussed.
Collapse
Affiliation(s)
- Sandra Demaria
- Department of Pathology, New York University School of Medicine, and NYU Cancer Institute, New York, New York 10016
- Department of Radiation Oncology, New York University School of Medicine, and NYU Cancer Institute, New York, New York 10016
| | - Karsten A. Pilones
- Department of Pathology, New York University School of Medicine, and NYU Cancer Institute, New York, New York 10016
| | - Claire Vanpouille-Box
- Department of Pathology, New York University School of Medicine, and NYU Cancer Institute, New York, New York 10016
| | - Encouse B. Golden
- Department of Radiation Oncology, New York University School of Medicine, and NYU Cancer Institute, New York, New York 10016
| | - Silvia C. Formenti
- Department of Radiation Oncology, New York University School of Medicine, and NYU Cancer Institute, New York, New York 10016
| |
Collapse
|
111
|
Verbrugge I, Gasparini A, Haynes NM, Hagekyriakou J, Galli M, Stewart TJ, Abrams SI, Yagita H, Verheij M, Johnstone RW, Borst J, Neefjes J. The Curative Outcome of Radioimmunotherapy in a Mouse Breast Cancer Model Relies on mTOR Signaling. Radiat Res 2014; 182:219-29. [DOI: 10.1667/rr13511.1] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/03/2022]
Affiliation(s)
- Inge Verbrugge
- Division of Immunology, The Netherlands Cancer Institute, Amsterdam, The Netherlands
| | - Alessia Gasparini
- Division of Radiotherapy, The Netherlands Cancer Institute, Amsterdam, The Netherlands
| | - Nicole M. Haynes
- Cancer Therapeutics Program, Peter MacCallum Cancer Centre, East Melbourne, Australia
| | - Jim Hagekyriakou
- Department of Physical Chemistry, Peter MacCallum Cancer Centre, East Melbourne, Australia
| | - Mara Galli
- Cancer Therapeutics Program, Peter MacCallum Cancer Centre, East Melbourne, Australia
| | - Trina J. Stewart
- Griffith Health Institute, School of Medical Sciences, Griffith University, Gold Coast, Australia
| | - Scott I. Abrams
- Department of Immunology, Roswell Park Cancer Institute, Buffalo, New York
| | - Hideo Yagita
- Department of Immunology, Juntendo University School of Medicine, Tokyo, Japan; and
| | - Marcel Verheij
- Division of Radiotherapy, The Netherlands Cancer Institute, Amsterdam, The Netherlands
| | - Ricky W. Johnstone
- Cancer Therapeutics Program, Peter MacCallum Cancer Centre, East Melbourne, Australia
| | - Jannie Borst
- Division of Immunology, The Netherlands Cancer Institute, Amsterdam, The Netherlands
| | - Jacques Neefjes
- Division of Cell Biology, The Netherlands Cancer Institute, Amsterdam, The Netherlands
| |
Collapse
|
112
|
Madireddi S, Eun SY, Lee SW, Nemčovičová I, Mehta AK, Zajonc DM, Nishi N, Niki T, Hirashima M, Croft M. Galectin-9 controls the therapeutic activity of 4-1BB-targeting antibodies. ACTA ACUST UNITED AC 2014; 211:1433-48. [PMID: 24958847 PMCID: PMC4076583 DOI: 10.1084/jem.20132687] [Citation(s) in RCA: 91] [Impact Index Per Article: 9.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Abstract
Biologics to TNF family receptors are prime candidates for therapy of immune disease. Whereas recent studies have highlighted a requirement for Fcγ receptors in enabling the activity of CD40, TRAILR, and GITR when engaged by antibodies, other TNFR molecules may be controlled by additional mechanisms. Antibodies to 4-1BB (CD137) are currently in clinical trials and can both augment immunity in cancer and promote regulatory T cells that inhibit autoimmune disease. We found that the action of agonist anti-4-1BB in suppressing autoimmune and allergic inflammation was completely dependent on Galectin-9 (Gal-9). Gal-9 directly bound to 4-1BB, in a site distinct from the binding site of antibodies and the natural ligand of 4-1BB, and Gal-9 facilitated 4-1BB aggregation, signaling, and functional activity in T cells, dendritic cells, and natural killer cells. Conservation of the Gal-9 interaction in humans has important implications for effective clinical targeting of 4-1BB and possibly other TNFR superfamily molecules.
Collapse
Affiliation(s)
- Shravan Madireddi
- Division of Immune Regulation and Division of Cell Biology, La Jolla Institute for Allergy and Immunology, La Jolla, CA 92037
| | - So-Young Eun
- Division of Immune Regulation and Division of Cell Biology, La Jolla Institute for Allergy and Immunology, La Jolla, CA 92037
| | - Seung-Woo Lee
- Division of Immune Regulation and Division of Cell Biology, La Jolla Institute for Allergy and Immunology, La Jolla, CA 92037
| | - Ivana Nemčovičová
- Division of Immune Regulation and Division of Cell Biology, La Jolla Institute for Allergy and Immunology, La Jolla, CA 92037
| | - Amit Kumar Mehta
- Division of Immune Regulation and Division of Cell Biology, La Jolla Institute for Allergy and Immunology, La Jolla, CA 92037
| | - Dirk M Zajonc
- Division of Immune Regulation and Division of Cell Biology, La Jolla Institute for Allergy and Immunology, La Jolla, CA 92037
| | - Nozomu Nishi
- Life Science Research Center; and Department of Immunology and Immunopathology, Faculty of Medicine; Kagawa University, Kagawa 761-0793, Japan
| | - Toshiro Niki
- Life Science Research Center; and Department of Immunology and Immunopathology, Faculty of Medicine; Kagawa University, Kagawa 761-0793, Japan GalPharma Co., Ltd., Kagawa 760-0301, Japan
| | - Mitsuomi Hirashima
- Life Science Research Center; and Department of Immunology and Immunopathology, Faculty of Medicine; Kagawa University, Kagawa 761-0793, Japan GalPharma Co., Ltd., Kagawa 760-0301, Japan
| | - Michael Croft
- Division of Immune Regulation and Division of Cell Biology, La Jolla Institute for Allergy and Immunology, La Jolla, CA 92037
| |
Collapse
|
113
|
Impact of tumour microenvironment and Fc receptors on the activity of immunomodulatory antibodies. Trends Immunol 2014; 35:290-8. [PMID: 24953012 DOI: 10.1016/j.it.2014.05.002] [Citation(s) in RCA: 80] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2014] [Revised: 05/06/2014] [Accepted: 05/12/2014] [Indexed: 12/20/2022]
Abstract
Immunomodulatory antibodies influence the direction and magnitude of immune responses against cancer. Significant efficacy has been demonstrated across multiple solid tumour types within clinical trials. Recent preclinical studies indicate that successful outcome relies upon mechanistic activity extending beyond simple receptor stimulation or blockade. In addition to blocking co-inhibitory signals in secondary lymphoid organs, cytotoxic T-lymphocyte antigen (CTLA)-4 antibodies mediate depletion of tumour-infiltrating regulatory T cells by antibody-dependent cellular cytotoxicity (ADCC). This mechanism appears to be common to other immunomodulatory antibodies including those targeting OX40 and glucocorticoid-induced TNFR-related protein (GITR). If verified in the human setting, these findings have significant implications for antibody design, biomarker discovery, and the development of synergistic combinatorial therapies.
Collapse
|
114
|
Bertolini M, Zilio F, Rossi A, Kleditzsch P, Emelianov VE, Gilhar A, Keren A, Meyer KC, Wang E, Funk W, McElwee K, Paus R. Abnormal interactions between perifollicular mast cells and CD8+ T-cells may contribute to the pathogenesis of alopecia areata. PLoS One 2014; 9:e94260. [PMID: 24832234 PMCID: PMC4022513 DOI: 10.1371/journal.pone.0094260] [Citation(s) in RCA: 99] [Impact Index Per Article: 9.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2013] [Accepted: 03/12/2014] [Indexed: 02/08/2023] Open
Abstract
Alopecia areata (AA) is a CD8+ T-cell dependent autoimmune disease of the hair follicle (HF) in which the collapse of HF immune privilege (IP) plays a key role. Mast cells (MCs) are crucial immunomodulatory cells implicated in the regulation of T cell-dependent immunity, IP, and hair growth. Therefore, we explored the role of MCs in AA pathogenesis, focusing on MC interactions with CD8+ T-cells in vivo, in both human and mouse skin with AA lesions. Quantitative (immuno-)histomorphometry revealed that the number, degranulation and proliferation of perifollicular MCs are significantly increased in human AA lesions compared to healthy or non-lesional control skin, most prominently in subacute AA. In AA patients, perifollicular MCs showed decreased TGFβ1 and IL-10 but increased tryptase immunoreactivity, suggesting that MCs switch from an immuno-inhibitory to a pro-inflammatory phenotype. This concept was supported by a decreased number of IL-10+ and PD-L1+ MCs, while OX40L+, CD30L+, 4–1BBL+ or ICAM-1+ MCs were increased in AA. Lesional AA-HFs also displayed significantly more peri- and intrafollicular- CD8+ T-cells as well as more physical MC/CD8+ T-cell contacts than healthy or non-lesional human control skin. During the interaction with CD8+ T-cells, AA MCs prominently expressed MHC class I and OX40L, and sometimes 4–1BBL or ICAM-1, suggesting that MC may present autoantigens to CD8+ T-cells and/or co-stimulatory signals. Abnormal MC numbers, activities, and interactions with CD8+ T-cells were also seen in the grafted C3H/HeJ mouse model of AA and in a new humanized mouse model for AA. These phenomenological in vivo data suggest the novel AA pathobiology concept that perifollicular MCs are skewed towards pro-inflammatory activities that facilitate cross-talk with CD8+ T-cells in this disease, thus contributing to triggering HF-IP collapse in AA. If confirmed, MCs and their CD8+ T-cell interactions could become a promising new therapeutic target in the future management of AA.
Collapse
Affiliation(s)
- Marta Bertolini
- Department of Dermatology, University of Lübeck, Lübeck, Germany
- Department of Dermatology, University of Münster, Münster, Germany
| | - Federica Zilio
- Department of Dermatology, University of Lübeck, Lübeck, Germany
| | - Alfredo Rossi
- Department of Internal Medicine and Medical Specialties, University “La Sapienza”, Rome, Italy
| | - Patrick Kleditzsch
- Department of Gynaecology and Obstetrics, University of Rostock, Rostock, Germany
| | - Vladimir E. Emelianov
- Department of Pharmacology, Clinical Pharmacology and Biochemistry, Chuvash State University Medical School, Cheboksary, Russia
| | - Amos Gilhar
- Laboratory for Skin Research, Rappaport Faculty of Medicine, Technion–Israel Institute of Technology, Haifa, Israel
- Flieman Medical Center, Haifa, Israel
| | - Aviad Keren
- Laboratory for Skin Research, Rappaport Faculty of Medicine, Technion–Israel Institute of Technology, Haifa, Israel
| | - Katja C. Meyer
- Department of Dermatology, University of Lübeck, Lübeck, Germany
| | - Eddy Wang
- Department of Dermatology and Skin Science, University of British Columbia, Vancouver, British Columbia, Canada
| | | | - Kevin McElwee
- Department of Dermatology and Skin Science, University of British Columbia, Vancouver, British Columbia, Canada
| | - Ralf Paus
- Department of Dermatology, University of Lübeck, Lübeck, Germany
- Department of Dermatology, University of Münster, Münster, Germany
- Institute for Inflammation and Repair, University of Manchester, Manchester, United Kingdom
- * E-mail:
| |
Collapse
|
115
|
Bae J, Prabhala R, Voskertchian A, Brown A, Maguire C, Richardson P, Dranoff G, Anderson KC, Munshi NC. A multiepitope of XBP1, CD138 and CS1 peptides induces myeloma-specific cytotoxic T lymphocytes in T cells of smoldering myeloma patients. Leukemia 2014; 29:218-29. [PMID: 24935722 PMCID: PMC4237716 DOI: 10.1038/leu.2014.159] [Citation(s) in RCA: 47] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2014] [Revised: 03/24/2014] [Accepted: 04/02/2014] [Indexed: 02/07/2023]
Abstract
We evaluated a cocktail of HLA-A2-specific peptides including heteroclitic XBP1 US184-192 (YISPWILAV), heteroclitic XBP1 SP367-375 (YLFPQLISV), native CD138260-268 (GLVGLIFAV) and native CS1239-247 (SLFVLGLFL), for their ability to elicit multipeptide-specific cytotoxic T lymphocytes (MP-CTLs) using T cells from smoldering multiple myeloma (SMM) patients. Our results demonstrate that MP-CTLs generated from SMM patients' T cells show effective anti-MM responses including CD137 (4-1BB) upregulation, CTL proliferation, interferon-γ production and degranulation (CD107a) in an HLA-A2-restricted and peptide-specific manner. Phenotypically, we observed increased total CD3(+)CD8(+) T cells (>80%) and cellular activation (CD69(+)) within the memory SMM MP-CTL (CD45RO(+)/CD3(+)CD8(+)) subset after repeated multipeptide stimulation. Importantly, SMM patients could be categorized into distinct groups by their level of MP-CTL expansion and antitumor activity. In high responders, the effector memory (CCR7(-)CD45RO(+)/CD3(+)CD8(+)) T-cell subset was enriched, whereas the remaining responders' CTL contained a higher frequency of the terminal effector (CCR7(-)CD45RO(-)/CD3(+)CD8(+)) subset. These results suggest that this multipeptide cocktail has the potential to induce effective and durable memory MP-CTL in SMM patients. Therefore, our findings provide the rationale for clinical evaluation of a therapeutic vaccine to prevent or delay progression of SMM to active disease.
Collapse
Affiliation(s)
- J Bae
- 1] Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA [2] Harvard Medical School, Boston, MA, USA
| | - R Prabhala
- 1] Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA [2] Harvard Medical School, Boston, MA, USA [3] VA Boston Healthcare System, Boston, MA, USA
| | - A Voskertchian
- Johns Hopkins School of Public Health, Baltimore, MD, USA
| | - A Brown
- 1] Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA [2] Harvard Medical School, Boston, MA, USA
| | - C Maguire
- Tufts University School of Medicine, Boston, MA, USA
| | - P Richardson
- 1] Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA [2] Harvard Medical School, Boston, MA, USA
| | - G Dranoff
- 1] Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA [2] Harvard Medical School, Boston, MA, USA
| | - K C Anderson
- 1] Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA [2] Harvard Medical School, Boston, MA, USA
| | - N C Munshi
- 1] Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA [2] Harvard Medical School, Boston, MA, USA [3] VA Boston Healthcare System, Boston, MA, USA
| |
Collapse
|
116
|
Westwood JA, Potdevin Hunnam TCU, Pegram HJ, Hicks RJ, Darcy PK, Kershaw MH. Routes of delivery for CpG and anti-CD137 for the treatment of orthotopic kidney tumors in mice. PLoS One 2014; 9:e95847. [PMID: 24788789 PMCID: PMC4008493 DOI: 10.1371/journal.pone.0095847] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2013] [Accepted: 04/01/2014] [Indexed: 12/24/2022] Open
Abstract
We have found previously that the tumor cell lines, Renca (a renal cancer) and MC38 (a colon tumor) which had been injected subcutaneously in mice, could be successfully treated with a combination therapy of an oligodeoxynucleotide (CpG1826) (injected intratumorally) and anti-CD137 antibody (injected intraperitoneally). Thus the combination treatment was expected to initiate a “danger” signal via TLR9 on immune cells, and the anti-CD137 was expected to further activate T cells. In the present study, we found that several other tumor types injected subcutaneously could also be successfully treated with this combination therapy. In addition, we wished to determine if the treatment could work as effectively in an orthotopic metastatic model, which is more physiologically relevant to cancer in humans. Renca was selected as we were familiar with injecting this orthotopically into the outer cortex of the kidney in mice, and it spontaneously metastasizes to lung and abdominal sites. We tested various routes of delivery of CpG combined with intraperitoneal delivery of anti-CD137. Orthotopic tumors were injected with CpG intratumorally, using ultrasound-guided delivery on multiple occasions, combined with anti-CD137 intraperitoneally. A reduction in primary tumor size was observed following intratumoral injection of CpG compared to other treatments. We found that there was a statistically significant increase in survival of mice with orthotopic Renca tumor following intratumoral injection of CpG. However, we determined that the most effective route of delivery of CpG was intravenous, which led to further significantly enhanced survival of mice when combined with anti-CD137 intraperitoneally, likely due to inhibition of metastatic disease. Our data supports future development of this combination therapy for cancer.
Collapse
Affiliation(s)
- Jennifer A. Westwood
- Division of Cancer Research, Peter MacCallum Cancer Centre, St Andrew's Place, East Melbourne, Australia
| | | | - Hollie J. Pegram
- Division of Cancer Research, Peter MacCallum Cancer Centre, St Andrew's Place, East Melbourne, Australia
| | - Rodney J. Hicks
- Centre for Cancer Imaging, Peter MacCallum Cancer Centre, St Andrew's Place, East Melbourne, Australia
| | - Phillip K. Darcy
- Division of Cancer Research, Peter MacCallum Cancer Centre, St Andrew's Place, East Melbourne, Australia
- Sir Peter MacCallum Department of Oncology, University of Melbourne, Parkville, Australia
- Department of Immunology, Monash University, Prahran, Australia
| | - Michael H. Kershaw
- Division of Cancer Research, Peter MacCallum Cancer Centre, St Andrew's Place, East Melbourne, Australia
- Sir Peter MacCallum Department of Oncology, University of Melbourne, Parkville, Australia
- Department of Immunology, Monash University, Prahran, Australia
- * E-mail:
| |
Collapse
|
117
|
Schaer DA, Hirschhorn-Cymerman D, Wolchok JD. Targeting tumor-necrosis factor receptor pathways for tumor immunotherapy. J Immunother Cancer 2014; 2:7. [PMID: 24855562 PMCID: PMC4030310 DOI: 10.1186/2051-1426-2-7] [Citation(s) in RCA: 90] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2014] [Accepted: 03/13/2014] [Indexed: 02/08/2023] Open
Abstract
With the success of ipilimumab and promise of programmed death-1 pathway-targeted agents, the field of tumor immunotherapy is expanding rapidly. Newer targets for clinical development include select members of the tumor necrosis factor receptor (TNFR) family. Agonist antibodies to these co-stimulatory molecules target both T and B cells, modulating T-cell activation and enhancing immune responses. In vitro and in vivo preclinical data have provided the basis for continued development of 4-1BB, OX40, glucocorticoid-induced TNFR-related gene, herpes virus entry mediator, and CD27 as potential therapies for patients with cancer. In this review, we summarize the immune response to tumors, consider preclinical and early clinical data on select TNFR family members, discuss potential translational challenges and suggest possible combination therapies with the aim of inducing durable antitumor responses.
Collapse
Affiliation(s)
- David A Schaer
- Swim Across America Laboratory, Immunology Program, Sloan-Kettering Institute for Cancer Research, New York, NY 10065, USA.,Current address: Department of Cancer Immunobiology, ImClone Systems, a wholly-owned subsidiary of Eli Lilly & Co, New York, NY 10016, USA
| | - Daniel Hirschhorn-Cymerman
- Swim Across America Laboratory, Immunology Program, Sloan-Kettering Institute for Cancer Research, New York, NY 10065, USA
| | - Jedd D Wolchok
- Swim Across America Laboratory, Immunology Program, Sloan-Kettering Institute for Cancer Research, New York, NY 10065, USA.,Weill Cornell Medical College, New York, NY 10065, USA.,Ludwig Collaborative Lab, New York, NY 10065, USA.,Ludwig Center for Cancer Immunotherapy at Memorial Sloan-Kettering Cancer Center, New York, NY 10065, USA
| |
Collapse
|
118
|
Song C, Sadashivaiah K, Furusawa A, Davila E, Tamada K, Banerjee A. Eomesodermin is required for antitumor immunity mediated by 4-1BB-agonist immunotherapy. Oncoimmunology 2014; 3:e27680. [PMID: 24790793 PMCID: PMC4002624 DOI: 10.4161/onci.27680] [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: 09/05/2013] [Revised: 12/29/2013] [Accepted: 12/29/2013] [Indexed: 12/30/2022] Open
Abstract
CD8+ T cells in progressing tumors frequently fail to mount an effective antitumor response often in association with the expression of inhibitory receptors, including programmed cell death-1 (PD-1) and lymphocyte-activation gene 3 (Lag3). Using a lymphoma tumor model, we demonstrate that tumor-infiltrating CD8+ T cells from growing tumors co-express inhibitory receptors and co-stimulatory receptors, including 4-1BB (TNFRSF9) as well as high levels of 2 transcription factors, Eomesodermin (Eomes) and T-bet (Tbx21), critical determinants of CD8+ T cell fate. Immunotherapy with an agonistic anti-4–1-BB antibody altered the ratio of Eomes to T-bet expression in tumor-infiltrating CD8+ T cells by increasing Eomes and decreasing T-bet expression. 4-1BB-agonist immunotherapy was also associated with downregulated expression of the inhibitory receptors PD-1 and Lag3 on tumor-infiltrating CD8+ T cells, a molecular phenotype associated with subsequent attenuation of tumor growth. Furthermore, 4-1BB-agonist immunotherapy failed to effect tumor progression in mice with Eomes deficient T cells. However, upon resumption of tumor growth, tumor-infiltrating CD8+ T cells from treated animals continued to express high levels of Eomes as well as elevated levels of the inhibitory receptors PD-1 and Lag3. Our data suggest that tumor-infiltrating CD8+ T cells are poised between activation and inhibition as dictated by expression of both co-stimulatory receptors and inhibitory receptors and demonstrate that T cell expression of Eomes is necessary, but not sufficient, for efficacious 4-1BB-agonist-mediated immunotherapy.
Collapse
Affiliation(s)
- Chang Song
- Program in Oncology; Greenebaum Cancer Center; Center for Stem Cell Research and Regenerative Medicine; Department of Medicine; University of Maryland School of Medicine; Baltimore, MD USA
| | - Kavitha Sadashivaiah
- Program in Oncology; Greenebaum Cancer Center; Center for Stem Cell Research and Regenerative Medicine; Department of Medicine; University of Maryland School of Medicine; Baltimore, MD USA
| | - Aki Furusawa
- Program in Oncology; Greenebaum Cancer Center; Center for Stem Cell Research and Regenerative Medicine; Department of Medicine; University of Maryland School of Medicine; Baltimore, MD USA
| | - Eduardo Davila
- Department of Otolaryngology; University of Maryland School of Medicine; Baltimore, MD USA
| | - Koji Tamada
- Department of Immunology and Cellular Signal Analysis; Yamaguchi University School of Medicine; Ube, Japan
| | - Arnob Banerjee
- Program in Oncology; Greenebaum Cancer Center; Center for Stem Cell Research and Regenerative Medicine; Department of Medicine; University of Maryland School of Medicine; Baltimore, MD USA
| |
Collapse
|
119
|
Németh T, Tóth A, Hamari Z, Falus A, Éder K, Vágvölgyi C, Guimaraes AJ, Nosanchuk JD, Gácser A. Transcriptome profile of the murine macrophage cell response to Candida parapsilosis. Fungal Genet Biol 2014; 65:48-56. [PMID: 24530442 DOI: 10.1016/j.fgb.2014.01.006] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2013] [Revised: 01/17/2014] [Accepted: 01/29/2014] [Indexed: 12/20/2022]
Abstract
Candida parapsilosis is a human fungal pathogen with increasing global significance. Understanding how macrophages respond to C. parapsilosis at the molecular level will facilitate the development of novel therapeutic paradigms. The complex response of murine macrophages to infection with C. parapsilosis was investigated at the level of gene expression using an Agilent mouse microarray. We identified 155 and 511 differentially regulated genes at 3 and 8h post-infection, respectively. Most of the upregulated genes encoded molecules involved in immune response and inflammation, transcription, signaling, apoptosis, cell cycle, electron transport and cell adhesion. Typical of the classically activated macrophages, there was significant upregulation of genes coordinating the production of inflammatory cytokines such as TNF, IL-1 and IL-15. Further, we used both primary murine macrophages and macrophages differentiated from human peripheral mononuclear cells to confirm the upregulation of the TNF-receptor family member TNFRSF9 that is associated with Th1 T-helper cell responses. Additionally, the microarray data indicate significant differences between the response to C. parapsilosis infection and that of C. albicans.
Collapse
Affiliation(s)
- Tibor Németh
- Department of Microbiology, University of Szeged, Közép fasor 52, H-6726 Szeged, Hungary
| | - Adél Tóth
- Department of Microbiology, University of Szeged, Közép fasor 52, H-6726 Szeged, Hungary
| | - Zsuzsanna Hamari
- Department of Microbiology, University of Szeged, Közép fasor 52, H-6726 Szeged, Hungary
| | - András Falus
- Department of Genetics, Cell- and Immunobiology, Semmelweis University, Nagyvárad tér 4, 1089 Budapest, Hungary
| | - Katalin Éder
- Department of Genetics, Cell- and Immunobiology, Semmelweis University, Nagyvárad tér 4, 1089 Budapest, Hungary
| | - Csaba Vágvölgyi
- Department of Microbiology, University of Szeged, Közép fasor 52, H-6726 Szeged, Hungary
| | - Allan J Guimaraes
- Department of Microbiology and Immunology, Albert Einstein College of Medicine, 1300 Morris Park Ave, New York, NY 10461, USA
| | - Joshua D Nosanchuk
- Department of Microbiology and Immunology, Albert Einstein College of Medicine, 1300 Morris Park Ave, New York, NY 10461, USA
| | - Attila Gácser
- Department of Microbiology, University of Szeged, Közép fasor 52, H-6726 Szeged, Hungary.
| |
Collapse
|
120
|
Yi L, Zhao Y, Wang X, Dai M, Hellström KE, Hellström I, Zhang H. Human and mouse CD137 have predominantly different binding CRDs to their respective ligands. PLoS One 2014; 9:e86337. [PMID: 24466035 PMCID: PMC3897701 DOI: 10.1371/journal.pone.0086337] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2013] [Accepted: 12/06/2013] [Indexed: 11/18/2022] Open
Abstract
Monoclonal antibodies (mAbs) to CD137 (a.k.a. 4-1BB) have anti-tumor efficacy in several animal models and have entered clinical trials in patients with advanced cancer. Importantly, anti-CD137 mAbs can also ameliorate autoimmunity in preclinical models. As an approach to better understand the action of agonistic and antagonistic anti-CD137 mAbs we have mapped the binding region of the CD137 ligand (CD137L) to human and mouse CD137. By investigating the binding of CD137L to cysteine rich domain II (CRDII )and CRDIII of CD137, we found that the binding interface was limited and differed between the two species in that mouse CD137L mainly combined with CRDII and human CD137L mainly combined with CRDIII.
Collapse
Affiliation(s)
- Ling Yi
- Department of Central Laboratory, Beijing Chest Hospital, Capital Medical University and Beijing Tuberculosis and Thoracic Tumor Research Institute, Beijing, People's Republic of China
| | - Yanlin Zhao
- Chinese Center for Disease Control and Prevention, Beijing, People's Republic of China
| | - Xiaojue Wang
- Department of Central Laboratory, Beijing Chest Hospital, Capital Medical University and Beijing Tuberculosis and Thoracic Tumor Research Institute, Beijing, People's Republic of China
| | - Min Dai
- Department of Pathology, Harborview Medical Center, University of Washington, Seattle, Washington, United States of America
| | - Karl Erik Hellström
- Department of Pathology, Harborview Medical Center, University of Washington, Seattle, Washington, United States of America
| | - Ingegerd Hellström
- Department of Pathology, Harborview Medical Center, University of Washington, Seattle, Washington, United States of America
| | - Hongtao Zhang
- Department of Central Laboratory, Beijing Chest Hospital, Capital Medical University and Beijing Tuberculosis and Thoracic Tumor Research Institute, Beijing, People's Republic of China
- * E-mail:
| |
Collapse
|
121
|
Black CM, Armstrong TD, Jaffee EM. Apoptosis-regulated low-avidity cancer-specific CD8(+) T cells can be rescued to eliminate HER2/neu-expressing tumors by costimulatory agonists in tolerized mice. Cancer Immunol Res 2014; 2:307-19. [PMID: 24764578 DOI: 10.1158/2326-6066.cir-13-0145] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
A major barrier to vaccines in cancer treatment is their failure to activate and maintain a complete cancer-specific CD8(+) effector T-cell repertoire. Low-avidity T cells are more likely to escape clonal deletion in the thymus when compared with high-avidity T cells, and therefore comprise the major population of effector T cells available for activation in patients with cancer. However, low-avidity T cells fail to traffic into the tumor microenvironment and function in eradicating tumor under optimal vaccination conditions as opposed to high-avidity T cells that escape clonal deletion and function in tumor killing. We used high- and low-avidity T-cell receptor transgenic CD8(+) T cells specific for the immunodominant epitope HER2/neu (RNEU420-429) to identify signaling pathways responsible for the inferior activity of the low-avidity T cells. Adoptive transfer of these cells into tumor-bearing vaccinated mice identified the members of apoptosis pathways that are upregulated in low-avidity T cells. The increased expression of proapoptotic proteins by low-avidity T cells promoted their own cell death and also that of other tumor-specific CD8(+) T cells within their local environment. Importantly, we show that this proapoptotic effect can be overcome by using a strong costimulatory signal that prevents the activation-induced cell death and enables the low-avidity T cells to traffic into the tumor and assist in tumor clearance. These findings identify new therapeutic opportunities for activating the most potent anticancer T-cell responses.
Collapse
Affiliation(s)
- Chelsea M Black
- Authors' Affiliations: Oncology, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | | | | |
Collapse
|
122
|
Kühnöl C, Herbarth M, Föll J, Staege MS, Kramm C. CD137 stimulation and p38 MAPK inhibition improve reactivity in an in vitro model of glioblastoma immunotherapy. Cancer Immunol Immunother 2013; 62:1797-809. [PMID: 24129764 PMCID: PMC11028552 DOI: 10.1007/s00262-013-1484-9] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2013] [Accepted: 10/03/2013] [Indexed: 10/26/2022]
Abstract
Dendritic cell vaccination has become an interesting option for cancer immunotherapy. Tumor-lysate-pulsed dendritic cells (DC) can prime naïve T cells and induce the regression of established tumors including gliomas as shown in various animal models. Despite hopeful results even in clinical studies, the outcome for many patients is still unsatisfying. In the present study, we tested the combination of tumor-lysate-pulsed dendritic cells (TPDC) with a monoclonal antibody against CD137, a monoclonal antibody against CD25 (daclizumab) and a specific p38 mitogen-activated protein kinase (p38 MAPK) inhibitor (SB203580) for improving immunostimulation in an in vitro model of immunotherapy for human gliomas. We observed a higher secretion of interferon gamma by TPDC-primed peripheral blood mononuclear cells (PBMC) that were incubated with an antibody against CD137 or the p38 MAPK inhibitor. In addition, we observed higher specific lysis of tumor cells after incubation of PBMC with the p38 MAPK inhibitor or the anti-CD137 antibody. In contrast, incubation of TPDC-primed PBMC with the anti-CD25 antibody did enhance neither interferon gamma secretion nor cellular cytotoxicity. Cell depletion experiments demonstrated that the immune reaction induced by TPDC is strongly dependent on CD4-positive and CD8-positive cells. Incubation of DC during maturation and antigen loading with the anti-CD137 antibody did not enhance cytotoxicity and interferon gamma secretion in comparison with application of the anti-CD137 antibody during priming. In conclusion, our data suggest that p38 MAPK inhibition and anti-CD137 antibodies can enhance the immune response against glioblastoma cells.
Collapse
MESH Headings
- Antibodies, Monoclonal, Humanized/pharmacology
- Blotting, Western
- Brain Neoplasms/immunology
- Brain Neoplasms/metabolism
- Brain Neoplasms/therapy
- Cell Proliferation
- Cells, Cultured
- Cytotoxicity, Immunologic
- Daclizumab
- Dendritic Cells/cytology
- Dendritic Cells/drug effects
- Dendritic Cells/immunology
- Enzyme Inhibitors/pharmacology
- Enzyme-Linked Immunosorbent Assay
- Flow Cytometry
- Glioblastoma/immunology
- Glioblastoma/metabolism
- Glioblastoma/therapy
- Humans
- Imidazoles/pharmacology
- Immunoglobulin G/pharmacology
- Immunosuppressive Agents/pharmacology
- Immunotherapy
- Interferon-gamma
- Interleukin-2 Receptor alpha Subunit/antagonists & inhibitors
- Interleukin-2 Receptor alpha Subunit/immunology
- Interleukin-2 Receptor alpha Subunit/metabolism
- Leukocytes, Mononuclear/cytology
- Leukocytes, Mononuclear/drug effects
- Leukocytes, Mononuclear/immunology
- Pyridines/pharmacology
- Tumor Necrosis Factor Receptor Superfamily, Member 9/immunology
- Tumor Necrosis Factor Receptor Superfamily, Member 9/metabolism
- p38 Mitogen-Activated Protein Kinases/antagonists & inhibitors
- p38 Mitogen-Activated Protein Kinases/immunology
- p38 Mitogen-Activated Protein Kinases/metabolism
Collapse
Affiliation(s)
- Caspar Kühnöl
- Department of Pediatrics, Martin-Luther-University Halle-Wittenberg, Ernst-Grube-Str. 40, 06097 Halle, Germany
| | - Monique Herbarth
- Department of Pediatrics, Martin-Luther-University Halle-Wittenberg, Ernst-Grube-Str. 40, 06097 Halle, Germany
| | - Jürgen Föll
- Department of Pediatrics and Juvenile Medicine, University Hospital Regensburg, 93053 Regensburg, Germany
| | - Martin S. Staege
- Department of Pediatrics, Martin-Luther-University Halle-Wittenberg, Ernst-Grube-Str. 40, 06097 Halle, Germany
| | - Christof Kramm
- Department of Pediatrics, Martin-Luther-University Halle-Wittenberg, Ernst-Grube-Str. 40, 06097 Halle, Germany
- Division of Pediatric Hematology and Oncology, Department of Pediatrics and Adolescent Medicine, University Medical Center Göttingen, 37075 Göttingen, Germany
| |
Collapse
|
123
|
Page DB, Postow MA, Callahan MK, Allison JP, Wolchok JD. Immune modulation in cancer with antibodies. Annu Rev Med 2013; 65:185-202. [PMID: 24188664 DOI: 10.1146/annurev-med-092012-112807] [Citation(s) in RCA: 401] [Impact Index Per Article: 36.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Ipilimumab is the prototypical immunomodulatory antibody, approved by the FDA in 2011 for advanced melanoma on the basis of survival benefit. Since that time, we have made significant strides in optimizing this therapy: we have characterized the spectrum of immune-related adverse events and learned how to mitigate them with treatment algorithms, discovered potential biomarkers of activity, and identified the potential synergy between checkpoint modulation and other therapeutic modalities. Recent phase I trials have established the efficacy and safety of next-generation checkpoint agents, including PD-1 and PD-L1 inhibitors, across multiple tumor types. Much work lies ahead in developing these next-generation checkpoint agents, testing them in combination, and determining how to integrate them into the treatment paradigms of various tumor types.
Collapse
Affiliation(s)
- David B Page
- Ludwig Center for Cancer Immunotherapy, Memorial Sloan-Kettering Cancer Center, New York, New York 10065; , , ,
| | | | | | | | | |
Collapse
|
124
|
Spranger S, Gajewski T. Rational combinations of immunotherapeutics that target discrete pathways. J Immunother Cancer 2013; 1:16. [PMID: 24829752 PMCID: PMC4019905 DOI: 10.1186/2051-1426-1-16] [Citation(s) in RCA: 57] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2013] [Accepted: 09/12/2013] [Indexed: 12/25/2022] Open
Abstract
An effective anti-tumor immune response requires the coordinated action of the innate and adaptive phases of the immune system. Critical processes include the activation of dendritic cells to present antigens, produce cytokines including type I interferons, and express multiple costimulatory ligands; induction of a productive T cell response within lymph nodes; migration of activated T cells to the tumor microenvironment in response to chemokines and homing receptor expression; and having effector T cells gain access to antigen-expressing tumor cells and maintain sufficient functionality to destroy them. However, tumors can become adept at escaping the immune response, developing multiple mechanisms to disrupt key processes. In general, tumors can be assigned into two different, major groups depending on whether the tumor there is an 'inflamed' or 'non-inflamed' tumor microenvironment. Improvements in our understanding of the interactions between the immune system and cancer have resulted in the development of various strategies to improve the immune-mediated control of tumors in both sub-groups. Categories of major immunotherapeutic intervention include methods to increase the frequency of tumor antigen-specific effector T cells in the circulation, strategies to block or uncouple a range of immune suppressive mechanisms within the tumor microenvironment, and tactics to induce de novo immune inflammation within the tumor microenvironment. The latter may be particularly important for eliciting immune recognition of non-inflamed tumor phenotypes. The premise put forth in this review is that synergistic therapeutic effects in vivo may be derived from combination therapies taken from distinct "bins" based on these mechanisms of action. Early data in both preclinical and some clinical studies provide support for this model. We also suggest that optimal application of these combinations may be aided by appropriate patient selection based on predictive biomarkers.
Collapse
Affiliation(s)
- Stefani Spranger
- Biological Sciences Division, Pathology, The University of Chicago, 929 E. 57th Street, GCIS W-423, Chicago, IL 60637, USA
| | - Thomas Gajewski
- Department of Pathology and Department of Medicine, Section of Hematology/Oncology, The University of Chicago, 5841 S. Maryland Ave., MC2115, Chicago, IL 60637, USA
| |
Collapse
|
125
|
Li SY, Liu Y. Immunotherapy of melanoma with the immune costimulatory monoclonal antibodies targeting CD137. Clin Pharmacol 2013; 5:47-53. [PMID: 24052693 PMCID: PMC3776567 DOI: 10.2147/cpaa.s46199] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Knowledge of how the immune system recognizes and attempts to control cancer growth and development has improved dramatically. The advent of immunotherapies for cancer has resulted in robust clinical responses and confirmed that the immune system can significantly inhibit tumor progression. Until recently, metastatic melanoma was a disease with limited treatment options and a poor prognosis. CD137 (also known as 4-1BB) a member of the tumor necrosis factor (TNF) receptor superfamily, is an activation-induced T cell costimulator molecule. Growing evidence indicates that anti-CD137 monoclonal antibodies possess strong antitumor properties, the result of their powerful capability to activate CD8+ T cells, to produce interferon (IFN)-γ, and to induce cytolytic markers. Combination therapy of anti-CD137 with other anticancer agents, such as radiation, has robust tumor-regressing abilities against nonimmunogenic or poorly immunogenic tumors. Of importance, targeting CD137 eliminates established tumors, and the fact that anti-CD137 therapy acts in concert with other anticancer agents and/or radiation therapy to eradicate nonimmunogenic and weakly immunogenic tumors is an additional benefit. Currently, BMS-663513, a humanized anti-CD137 antibody, is in clinical trials in patients with solid tumors, including melanoma, renal carcinoma, ovarian cancer, and B-cell malignancies. In this review, we discuss the basis of the therapeutic potential of targeting CD137 in cancer treatment, focusing in particular, on BMS-663513 as an immune costimulatory monoclonal antibody for melanoma immunotherapy.
Collapse
Affiliation(s)
- Shi-Yan Li
- Cancer Research Institute, Scott and white Healthcare, Temple, TX, USA
| | | |
Collapse
|
126
|
Escuin-Ordinas H, Elliott MW, Atefi M, Lee M, Ng C, Wei L, Comin-Anduix B, Montecino-Rodriguez E, Avramis E, Radu C, Sharp LL, Ribas A. PET imaging to non-invasively study immune activation leading to antitumor responses with a 4-1BB agonistic antibody. J Immunother Cancer 2013; 1:14. [PMID: 24829750 PMCID: PMC4019904 DOI: 10.1186/2051-1426-1-14] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2013] [Accepted: 08/07/2013] [Indexed: 11/20/2022] Open
Abstract
Background Molecular imaging with positron emission tomography (PET) may allow the non-invasive study of the pharmacodynamic effects of agonistic monoclonal antibodies (mAb) to 4-1BB (CD137). 4-1BB is a member of the tumor necrosis factor family expressed on activated T cells and other immune cells, and activating 4-1BB antibodies are being tested for the treatment of patients with advanced cancers. Methods We studied the antitumor activity of 4-1BB mAb therapy using [18 F]-labeled fluoro-2-deoxy-2-D-glucose ([18 F]FDG) microPET scanning in a mouse model of colon cancer. Results of microPET imaging were correlated with morphological changes in tumors, draining lymph nodes as well as cell subset uptake of the metabolic PET tracer in vitro. Results The administration of 4-1BB mAb to Balb/c mice induced reproducible CT26 tumor regressions and improved survival; complete tumor shrinkage was achieved in the majority of mice. There was markedly increased [18 F]FDG signal at the tumor site and draining lymph nodes. In a metabolic probe in vitro uptake assay, there was an 8-fold increase in uptake of [3H]DDG in leukocytes extracted from tumors and draining lymph nodes of mice treated with 4-1BB mAb compared to untreated mice, supporting the in vivo PET data. Conclusion Increased uptake of [18 F]FDG by PET scans visualizes 4-1BB agonistic antibody-induced antitumor immune responses and can be used as a pharmacodynamic readout to guide the development of this class of antibodies in the clinic.
Collapse
Affiliation(s)
- Helena Escuin-Ordinas
- Department of Medicine (Division of Hematology-Oncology) at David Geffen School of Medicine, University of California Los Angeles (UCLA), Los Angeles, USA
| | - Mark W Elliott
- Pfizer Worldwide Research and Development, Oncology Research Unit, San Diego, CA, USA
| | - Mohammad Atefi
- Department of Medicine (Division of Hematology-Oncology) at David Geffen School of Medicine, University of California Los Angeles (UCLA), Los Angeles, USA
| | - Michelle Lee
- Pfizer Worldwide Research and Development, Oncology Research Unit, San Diego, CA, USA
| | - Charles Ng
- Department of Medicine (Division of Hematology-Oncology) at David Geffen School of Medicine, University of California Los Angeles (UCLA), Los Angeles, USA
| | - Liu Wei
- Ahmanson Translational Imaging Division, Department of Molecular and Medical Pharmacology, UCLA, Los Angeles, USA
| | - Begoña Comin-Anduix
- Department of Surgery (Division of Surgical-Oncology), UCLA, Los Angeles, USA ; Jonsson Comprehensive Cancer Center (JCCC), Los Angeles, USA
| | | | - Earl Avramis
- Department of Surgery (Division of Surgical-Oncology), UCLA, Los Angeles, USA
| | - Caius Radu
- Ahmanson Translational Imaging Division, Department of Molecular and Medical Pharmacology, UCLA, Los Angeles, USA ; Jonsson Comprehensive Cancer Center (JCCC), Los Angeles, USA
| | - Leslie L Sharp
- Pfizer Worldwide Research and Development, Oncology Research Unit, San Diego, CA, USA ; Current address: Genomics Institute of the Novartis Research Foundation, 10675 John Jay Hopkins Dr., San Diego, CA 92121, USA
| | - Antoni Ribas
- Department of Medicine (Division of Hematology-Oncology) at David Geffen School of Medicine, University of California Los Angeles (UCLA), Los Angeles, USA ; Ahmanson Translational Imaging Division, Department of Molecular and Medical Pharmacology, UCLA, Los Angeles, USA ; Jonsson Comprehensive Cancer Center (JCCC), Los Angeles, USA ; Department of Medicine, Division of Hematology-Oncology, 11-934 Factor Building, Jonsson Comprehensive Cancer Center at UCLA, 10833 Le Conte Avenue, Los Angeles, CA 90095-1782, USA
| |
Collapse
|
127
|
Martínez Gómez JM, Koh VHQ, Yan B, Lin W, Ang MLT, Rahim SZZ, Pethe K, Schwarz H, Alonso S. Role of the CD137 ligand (CD137L) signaling pathway during Mycobacterium tuberculosis infection. Immunobiology 2013; 219:78-86. [PMID: 24091276 DOI: 10.1016/j.imbio.2013.08.009] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2013] [Revised: 08/06/2013] [Accepted: 08/12/2013] [Indexed: 02/01/2023]
Abstract
The role of the CD137-CD137 ligand (CD137L) signaling pathway in T cell co-stimulation has been well established. Dysregulated CD137 or CD137L stimulation can lead to pathological conditions such as inflammatory diseases or cancer. However, the contribution of CD137-CD137L interaction to the control of infectious diseases has not been extensively studied, with the few available reports focusing mainly on viral infections. Here we investigated the role of the CD137-CD137L interactions during Mycobacterium tuberculosis infection. Using CD137L-deficient mice, we found that absence of the CD137L-mediated signaling pathway during M. tuberculosis infection resulted in delayed activation of CD4(+) T cells in the draining lymph nodes. This finding was supported by an in vitro mixed lymphocyte reaction assay that revealed impaired priming of T cells by CD137L-deficient dendritic cells upon mycobacterial infection. In addition, greater numbers of CD4(+) T cells and antigen presenting cells were measured in the lungs of CD137L-deficient mice. Strikingly, the lung cytokine production profile was profoundly altered in M. tuberculosis-infected CD137L-deficient mice with lower levels of TNF-α, IL-12 and IL-6 and elevated concentrations of IL-17 compared to their wild type counterparts. However and surprisingly, these tangible immunological disorders translated only into a mild and transient increase in the bacterial loads and a higher number of granulomatous lesions with impaired architecture in the lungs of the CD137L-deficient infected mice. Together, while our data support the engagement of the CD137L signaling pathway during M. tuberculosis infection, they underscore the functional redundancy and robustness of the host defense arsenal deployed against mycobacterial infection.
Collapse
Affiliation(s)
- Julia María Martínez Gómez
- Department of Microbiology, Yong Loo Lin School of Medicine, Life Sciences Institute, National University of Singapore, Singapore; Immunology Programme, Yong Loo Lin School of Medicine, Life Sciences Institute, National University of Singapore, Singapore
| | | | | | | | | | | | | | | | | |
Collapse
|
128
|
Zikich D, Schachter J, Besser MJ. Immunotherapy for the management of advanced melanoma: the next steps. Am J Clin Dermatol 2013; 14:261-72. [PMID: 23516145 DOI: 10.1007/s40257-013-0013-0] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023]
Abstract
Melanoma is an immunogenic tumor that can induce a natural immune response. A number of immunotherapy-based approaches have been developed over the past decades, and certain degrees of effectiveness were achieved by the use of cytokines, adoptive cell transfer and T-cell immune modulators. Currently, interleukin-2 and the immune stimulatory antibody, ipilimumab, are the only two approved immunotherapies for metastatic melanoma, but various new therapies are in promising developmental stages. This comprehensive review will discuss the latest achievements of immunotherapy and emerging directions for the management of advanced melanoma.
Collapse
Affiliation(s)
- Dragoslav Zikich
- Ella Institute for Melanoma, Sheba Medical Center, 52621 Ramat-Gan, Israel
| | | | | |
Collapse
|
129
|
Daniel-Meshulam I, Horovitz-Fried M, Cohen CJ. Enhanced antitumor activity mediated by human 4-1BB-engineered T cells. Int J Cancer 2013; 133:2903-13. [PMID: 23754772 DOI: 10.1002/ijc.28320] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2013] [Accepted: 05/17/2013] [Indexed: 01/05/2023]
Abstract
4-1BB (CD137) is a costimulatory molecule transiently expressed on the T-cell surface after TCR engagement, whereas its ligand 4-1BBL can be found on professional antigen-presenting cells, but more importantly, also on tumor cells. As the role of the 4-1BB/4-1BBL pathway has emerged central to CD8(+) T-cell responses and survival, we sought to test its relevance in the context of genetically modified human T cells. To that end, T cells purified from healthy donors and from vaccinated-melanoma patients were transduced to express high levels of constitutive 4-1BB. 4-1BB-transduced T cells were cocultured with melanoma tumor lines and exhibited enhanced cytokine secretion, upregulation of activation markers as well as increased cytotoxicity in a chick-chorioallantoic membrane model of human melanoma tumors. In addition, these cells expanded and proliferated at a higher rate, expressed heightened levels of the antiapoptotic molecule Bcl(XL) and were also relatively insensitive to immunosuppression mediated by transforming growth factor-β, compared to control cells. We also show that 4-1BBL expression on the target cell is essential to 4-1BB-mediated functional improvement. Overall, we conclude that the modification of human T cells with 4-1BB yields enhanced antitumor function which may have important applications in therapies based on the genetic modification of patient lymphocytes.
Collapse
Affiliation(s)
- Inbal Daniel-Meshulam
- Laboratory of Tumor Immunology and Immunotherapy, The Goodman Faculty of Life Sciences, Bar-Ilan University, Ramat, Gan, Israel
| | | | | |
Collapse
|
130
|
Lee DY, Choi BK, Lee DG, Kim YH, Kim CH, Lee SJ, Kwon BS. 4-1BB signaling activates the t cell factor 1 effector/β-catenin pathway with delayed kinetics via ERK signaling and delayed PI3K/AKT activation to promote the proliferation of CD8+ T Cells. PLoS One 2013; 8:e69677. [PMID: 23874982 PMCID: PMC3708905 DOI: 10.1371/journal.pone.0069677] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2012] [Accepted: 06/13/2013] [Indexed: 01/06/2023] Open
Abstract
4-1BB (CD137), an inducible costimulatory molecule, strongly enhances the proliferation and effector function of CD8(+) T cells. Since the serine/threonine kinase, glycogen synthase kinase-3 (GSK-3), is involved in a variety of signaling pathways of cellular proliferation, migration, immune responses, and apoptosis, we examined whether 4-1BB signaling activates GSK-3/β-catenin signaling and downstream transcription factors to enhance the proliferation of CD8(+) T cells. 4-1BB signaling induces rapid activation of ERK and IκB degradation, and shows delayed activation of AKT at 24 h post 4-1BB stimulation on anti-CD3 activated T cells. ERK and AKT signals were required for sustained β-catenin levels by inactivating GSK-3, which was also observed with delayed kinetics after 4-1BB stimulation. As a transcriptional partner of β-catenin, 4-1BB signaling decreased levels of FOXO1 and increased levels of stimulatory TCF1 in CD8(+) T cells at 2-3 days but not at early time points after 4-1BB engagement. The enhanced proliferation of CD8(+) T cells due to 4-1BB signaling was completely abolished by treatment with the TCF1/β-catenin inhibitor quercetin. These results show that 4-1BB signaling enhances the proliferation of activated CD8(+) T cells by activating the TCF1/β-catenin axis via the PI3K/AKT/ERK pathway. As effects of 4-1BB on AKT, FOXO1, β-catenin and GSK-3β showed delayed kinetics it is likely that an intervening molecule induced by 4-1BB and ERK signaling in activated T cells is responsible for these effects. These effects were observed on CD8(+) but not on CD4(+) T cells. Moreover, 4-1BB appeared to be unique among several TNFRs tested in inducing increase in stimulatory over inhibitory TCF-1.
Collapse
Affiliation(s)
- Do Y. Lee
- Cancer Immunology Branch, Division of Cancer Biology, National Cancer Center, Ilsan, Goyang, Gyeonggi-do, Korea
| | - Beom K. Choi
- Cancer Immunology Branch, Division of Cancer Biology, National Cancer Center, Ilsan, Goyang, Gyeonggi-do, Korea
| | - Don G. Lee
- Cancer Immunology Branch, Division of Cancer Biology, National Cancer Center, Ilsan, Goyang, Gyeonggi-do, Korea
| | - Young H. Kim
- Immune Cell Production Unit, Program for Immunotherapeutic Research, National Cancer Center, Ilsan, Goyang, Gyeonggi-do, Korea
| | - Chang H. Kim
- Cancer Immunology Branch, Division of Cancer Biology, National Cancer Center, Ilsan, Goyang, Gyeonggi-do, Korea
| | - Seung J. Lee
- Cancer Immunology Branch, Division of Cancer Biology, National Cancer Center, Ilsan, Goyang, Gyeonggi-do, Korea
| | - Byoung S. Kwon
- Cancer Immunology Branch, Division of Cancer Biology, National Cancer Center, Ilsan, Goyang, Gyeonggi-do, Korea
- Section of Clinical Immunology, Allergy, and Rheumatology, Department of Medicine, Tulane University Health Sciences Center, New Orleans, Louisiana, United States of America
- * E-mail:
| |
Collapse
|
131
|
So T, Croft M. Regulation of PI-3-Kinase and Akt Signaling in T Lymphocytes and Other Cells by TNFR Family Molecules. Front Immunol 2013; 4:139. [PMID: 23760533 PMCID: PMC3675380 DOI: 10.3389/fimmu.2013.00139] [Citation(s) in RCA: 88] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2013] [Accepted: 05/25/2013] [Indexed: 12/22/2022] Open
Abstract
Activation of phosphoinositide 3-kinase (PI3K) and Akt (protein kinase B) is a common response triggered by a range of membrane-bound receptors on many cell types. In T lymphocytes, the PI3K-Akt pathway promotes clonal expansion, differentiation, and survival of effector cells and suppresses the generation of regulatory T cells. PI3K activation is tightly controlled by signals through the T cell receptor (TCR) and the co-stimulatory receptor CD28, however sustained and periodic signals from additional co-receptors are now being recognized as critical contributors to the activation of this pathway. Accumulating evidence suggests that many members of the Tumor Necrosis Factor receptor (TNFR) superfamily, TNFR2 (TNFRSF1B), OX40 (TNFRSF4), 4-1BB (TNFRSF9), HVEM (TNFRSF14), and DR3 (TNFRSF25), that are constitutive or inducible on T cells, can directly or indirectly promote activity in the PI3K-Akt pathway. We discuss recent data which suggests that ligation of one TNFR family molecule organizes a signalosome, via TNFR-associated factor (TRAF) adapter proteins in T cell membrane lipid microdomains, that results in the subsequent accumulation of highly concentrated depots of PI3K and Akt in close proximity to TCR signaling units. We propose this may be a generalizable mechanism applicable to other TNFR family molecules that will result in a quantitative contribution of these signalosomes to enhancing and sustaining PI3K and Akt activation triggered by the TCR. We also review data that other TNFR molecules, such as CD40 (TNFRSF5), RANK (TNFRSF11A), FN14 (TNFRSF12A), TACI (TNFRSF13B), BAFFR (TNFRSF13C), and NGFR (TNFRSF16), contribute to the activation of this pathway in diverse cell types through a similar ability to recruit PI3K or Akt into their signaling complexes.
Collapse
Affiliation(s)
- Takanori So
- Department of Microbiology and Immunology, Tohoku University Graduate School of Medicine , Sendai , Japan
| | | |
Collapse
|
132
|
Crivello P, Lauterbach N, Zito L, Sizzano F, Toffalori C, Marcon J, Curci L, Mulder A, Wieten L, Zino E, Voorter CEM, Tilanus MGJ, Fleischhauer K. Effects of transmembrane region variability on cell surface expression and allorecognition of HLA-DP3. Hum Immunol 2013; 74:970-7. [PMID: 23619468 DOI: 10.1016/j.humimm.2013.04.014] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2012] [Revised: 03/06/2013] [Accepted: 04/12/2013] [Indexed: 12/01/2022]
Abstract
The functional relevance of polymorphisms outside the peptide binding groove of HLA molecules is poorly understood. Here we have addressed this issue by studying HLA-DP3, a common antigen relevant for functional matching algorithms of unrelated hematopoietic stem cell transplantation (HSCT) encoded by two transmembrane (TM) region variants, DPB1(*)03:01 and DPB1(*)104:01. The two HLA-DP3 variants were found at a overall allelic frequency of 10.4% in 201 volunteer stem cell donors, at a ratio of 4.2:1. No significant differences were observed in cell surface expression levels of the two variants on B lymphoblastoid cell lines (BLCL), primary B cells or monocytes. Three different alloreactive T cell lines or clones showed similar levels of activation marker CD107a and/or CD137 upregulation in response to HLA-DP3 encoded by DPB1(*)03:01 and DPB1(*)104:01, either endogenously on BLCL or after lentiveral-vector mediated transfer into the same cellular background. These data provide, for the first time, direct evidence for a limited functional role of a TM region polymorphism on expression and allorecognition of HLA-DP3 and are compatible with the notion that the two variants can be considered as a single functional entity for unrelated stem cell donor selection.
Collapse
Affiliation(s)
- Pietro Crivello
- Unit of Molecular and Functional Immunogenetics, San Raffaele Scientific Institute, Milan, Italy
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
133
|
Noncanonical roles of the immune system in eliciting oncogene addiction. Curr Opin Immunol 2013; 25:246-58. [PMID: 23571026 DOI: 10.1016/j.coi.2013.02.003] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2012] [Revised: 02/07/2013] [Accepted: 02/08/2013] [Indexed: 02/08/2023]
Abstract
Cancer is highly complex. The magnitude of this complexity makes it highly surprising that even the brief suppression of an oncogene can sometimes result in rapid and sustained tumor regression, illustrating that cancers can be 'oncogene addicted' [1-10]. The essential implication is that oncogenes may not only fuel the initiation of tumorigenesis, but in some cases must be excessively activated to maintain a neoplastic state [11]. Oncogene suppression acutely restores normal physiological programs that effectively overrides secondary genetic events and a cancer collapses [12,13]. Oncogene addiction is the description of the dramatic and sustained regression of some cancers upon the specific inactivation of a single oncogene [1-13,14(••),15,16(••)], that can occur through tumor intrinsic [1,2,4,12], but also host immune mechanisms [17-23]. Notably, oncogene inactivation elicits a host immune response that involves specific immune effectors and cytokines that facilitate a remodeling of the tumor microenvironment including the shut down of angiogenesis and the induction of cellular senescence of tumor cells [16(••)]. Hence, immune effectors are not only critically involved in tumor prevention, initiation [17-19], and progression [20], but also appear to be essential to tumor regression upon oncogene inactivation [21,22(••),23(••)]. Understanding how the inactivation of an oncogene elicits a systemic signal in the host that prompts a deconstruction of a tumor could have important implications. The combination of oncogene-targeted therapy together with immunomodulatory therapy may be ideal for the development of both robust tumor intrinsic and immunological responses, effectively leading to sustained tumor regression.
Collapse
|
134
|
Ye J, Li L, Zhang Y, Zhang X, Ren D, Chen W. Recombinant Salmonella-based 4-1BBL vaccine enhances T cell immunity and inhibits the development of colorectal cancer in rats: in vivo effects of vaccine containing 4-1BBL. J Biomed Sci 2013; 20:8. [PMID: 23413971 PMCID: PMC3605352 DOI: 10.1186/1423-0127-20-8] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2012] [Accepted: 02/08/2013] [Indexed: 11/22/2022] Open
Abstract
Background Immunotherapy with vaccines is attractive for the treatment of cancer. This study is aimed at determining the effect of recombinant Salmonella (SL3261)-based 4-1BB ligand (4-1BBL) vaccine on the development of colorectal cancers and the potential immune mechanisms in rats. Results In comparison with that in the PBS group, similar levels of 4-1BBL expression, the frequency of T cells, IFN-γ responses, and comparable numbers of tumors were detected in the SL3261 and SL3261C groups of rats. In contrast, significantly fewer numbers of tumors, increased levels of 4-1BBL expression in the spleens and colorectal tissues, higher frequency of peripheral blood and splenic CD3+CD25+ T cells, and stronger splenic T cell IFN-γ responses were detected in the SL3261R group of rats. Conclusion Our results indicated that vaccination with recombinant attenuated Salmonella harboring the 4-1BBL gene efficiently enhanced T cell immunity and inhibited the development of carcinogen-induced colorectal cancers in rats.
Collapse
Affiliation(s)
- Jianxin Ye
- Department of Gastroenterology, the First Affiliated Hospital of Soochow University, Suzhou 215006, China
| | | | | | | | | | | |
Collapse
|
135
|
Youlin K, Li Z, Xin G, Mingchao X, Xiuheng L, Xiaodong W. Enhanced function of cytotoxic T lymphocytes induced by dendritic cells modified with truncated PSMA and 4-1BBL. Hum Vaccin Immunother 2013; 9:766-72. [PMID: 23295983 DOI: 10.4161/hv.23116] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/25/2023] Open
Abstract
Interactions between costimulatory molecules and their receptors are vital for Ag-presenting dendritic cells (DCs) to initiate T cells activation, expansion and their antitumor immune responses. Augmentation of costimulatory signal due to the interaction of DCs and T cells may amplify, sustain and drive diversity of cytotoxic T lymphocytes (CTLs) and consequently enhance the antitumor response. 4-1BBL/4-1BB is such a pair of costimulatory ligand and receptor, playing an important role in the co-stimulation of CTLs. Previously, we demonstrated that DCs transduced with recombinant adenovirus encoding truncated PSMA (tPSMA) and m4-1BBL could induce prostate cancer regression in mouse models. In the present study, we further explored the adjuvant role of 4-1BBL in modulating CTLs activation induced by tPSMA gene-pulsed DCs. The apoptosis and cytotoxicity against tPSMA expressing RM-1 cells of CTLs were determined. Results showed that tPSMA gene-pulsed DCs effectively induced T lymphocyte activation and cytotoxicity, which was enhanced by upregulated expression of 4-1BBL, displaying better cell viability, lower CTLs apoptosis, higher expression anti-apoptotic protein of Bcl-xL and phosphorylation of P38, enhanced NF-κB activation, as well as more IFN-γ production. These results demonstrated that 4-1BBL may play a significant role in the co-stimulation pathway for Ag-presenting DCs-mediated CTLs activity, which might be a beneficial adjuvant factor for DCs-based cancer immunotherapy.
Collapse
Affiliation(s)
- Kuang Youlin
- Department of Urology; The First Affiliated Hospital; Chongqing Medical University; Chongqing, P.R. China
| | | | | | | | | | | |
Collapse
|
136
|
Barao I. The TNF receptor-ligands 4-1BB-4-1BBL and GITR-GITRL in NK cell responses. Front Immunol 2013; 3:402. [PMID: 23316193 PMCID: PMC3539674 DOI: 10.3389/fimmu.2012.00402] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2012] [Accepted: 12/14/2012] [Indexed: 11/19/2022] Open
Abstract
Interactions between several tumor necrosis factor (TNF)-TNF receptor (TNFR) superfamily members that are expressed by T cells and natural killer (NK) cells and various other cell types modulate immune responses. This review summarizes the current understanding of how the TNF ligand-TNFR interactions 4-1BBL with 4-1BB, and GITRL with glucocorticoid-induced TNFR-related (GITR) regulate NK cell mediated antitumor responses and discuss its therapeutic implications.
Collapse
Affiliation(s)
- Isabel Barao
- Department of Microbiology and Immunology, University of Nevada, Reno Reno, NV, USA
| |
Collapse
|
137
|
Melief CJM. Selective activation of oxygen-deprived tumor-infiltrating lymphocytes through local intratumoral delivery of CD137 monoclonal antibodies. Cancer Discov 2012; 2:586-7. [PMID: 22787088 DOI: 10.1158/2159-8290.cd-12-0229] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Hypoxia-inducing transcription factor-1α (HIF-1α) in hypoxic tumors induces the TNF receptor family member CD137 on tumor-infiltrating lymphocytes. This can be exploited for intratumoral low-dose injection of effective systemic immunotherapy with agonist CD137-specific monoclonal antibodies that induce circulation of systemic tumor-specific effector T cells capable of eradicating distant metastases.
Collapse
Affiliation(s)
- Cornelis J M Melief
- Immune System Activation, Leiden University Medical Center, Leiden, The Netherlands.
| |
Collapse
|