1
|
Redmond WL. Challenges and opportunities in the development of combination immunotherapy with OX40 agonists. Expert Opin Biol Ther 2023; 23:901-912. [PMID: 37587644 PMCID: PMC10530613 DOI: 10.1080/14712598.2023.2249396] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2023] [Accepted: 08/15/2023] [Indexed: 08/18/2023]
Abstract
INTRODUCTION Costimulatory members of the tumor necrosis factor receptor family, such as OX40 (CD134), provide essential survival and differentiation signals that enhance T cell function. Specifically, OX40 (CD134) agonists stimulate potent anti-tumor immunity in a variety of preclinical models but their therapeutic impact in patients with advanced malignancies has been limited thus far. AREAS COVERED In this review, we discuss the current state of combination immunotherapy with OX40 agonists including preclinical studies and recent clinical trials. We also discuss the strengths and limitations of these approaches and provide insight into alternatives that may help enhance the efficacy of combination OX40 agonist immunotherapy. EXPERT OPINION OX40 agonist immunotherapy has not yet demonstrated significant clinical activity as a monotherapy or in combination with immune checkpoint blockade (ICB), likely due to several factors including the timing of administration, drug potency, and selection of agents for combination therapy clinical trials. We believe that careful consideration of the biological mechanisms regulating OX40 expression and function may help inform new approaches, particularly in combination with novel agents, capable of increasing the therapeutic efficacy of this approach.
Collapse
Affiliation(s)
- William L Redmond
- Earle A. Chiles Research Institute, Providence Cancer Institute, 4805 NE Glisan St., 2N35, Portland, OR, 97213
| |
Collapse
|
2
|
Melero I, Sanmamed MF, Glez-Vaz J, Luri-Rey C, Wang J, Chen L. CD137 (4-1BB)-Based Cancer Immunotherapy on Its 25th Anniversary. Cancer Discov 2023; 13:552-569. [PMID: 36576322 DOI: 10.1158/2159-8290.cd-22-1029] [Citation(s) in RCA: 27] [Impact Index Per Article: 13.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2022] [Revised: 10/28/2022] [Accepted: 11/21/2022] [Indexed: 12/29/2022]
Abstract
Twenty-five years ago, we reported that agonist anti-CD137 monoclonal antibodies eradicated transplanted mouse tumors because of enhanced CD8+ T-cell antitumor immunity. Mouse models indicated that anti-CD137 agonist antibodies synergized with various other therapies. In the clinic, the agonist antibody urelumab showed evidence for single-agent activity against melanoma and non-Hodgkin lymphoma but caused severe liver inflammation in a fraction of the patients. CD137's signaling domain is included in approved chimeric antigen receptors conferring persistence and efficacy. A new wave of CD137 agonists targeting tumors, mainly based on bispecific constructs, are in early-phase trials and are showing promising safety and clinical activity. SIGNIFICANCE CD137 (4-1BB) is a costimulatory receptor of T and natural killer lymphocytes whose activity can be exploited in cancer immunotherapy strategies as discovered 25 years ago. Following initial attempts that met unacceptable toxicity, new waves of constructs acting agonistically on CD137 are being developed in patients, offering signs of clinical and pharmacodynamic activity with tolerable safety profiles.
Collapse
Affiliation(s)
- Ignacio Melero
- Program of Immunology and Immunotherapy, Center for Applied Medical Research (CIMA), Pamplona, Spain
- Navarra Institute for Health Research (IdiSNA), Pamplona, Spain
- Departments of Immunology-Immunotherapy and Oncology, Clínica Universidad de Navarra, Pamplona, Spain
- Centro de Investigación Biomédica en Red de Cáncer (CIBERONC), Madrid, Spain
| | - Miguel F Sanmamed
- Program of Immunology and Immunotherapy, Center for Applied Medical Research (CIMA), Pamplona, Spain
- Navarra Institute for Health Research (IdiSNA), Pamplona, Spain
- Departments of Immunology-Immunotherapy and Oncology, Clínica Universidad de Navarra, Pamplona, Spain
- Centro de Investigación Biomédica en Red de Cáncer (CIBERONC), Madrid, Spain
| | - Javier Glez-Vaz
- Program of Immunology and Immunotherapy, Center for Applied Medical Research (CIMA), Pamplona, Spain
- Navarra Institute for Health Research (IdiSNA), Pamplona, Spain
| | - Carlos Luri-Rey
- Program of Immunology and Immunotherapy, Center for Applied Medical Research (CIMA), Pamplona, Spain
- Navarra Institute for Health Research (IdiSNA), Pamplona, Spain
| | - Jun Wang
- Department of Pathology, New York University Grossman School of Medicine, New York, New York
| | - Lieping Chen
- Department of Immunobiology, Yale University, New Haven, Connecticut
| |
Collapse
|
3
|
Santiago-Sánchez GS, Hodge JW, Fabian KP. Tipping the scales: Immunotherapeutic strategies that disrupt immunosuppression and promote immune activation. Front Immunol 2022; 13:993624. [PMID: 36159809 PMCID: PMC9492957 DOI: 10.3389/fimmu.2022.993624] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2022] [Accepted: 08/17/2022] [Indexed: 11/13/2022] Open
Abstract
Immunotherapy has emerged as an effective therapeutic approach for several cancer types. However, only a subset of patients exhibits a durable response due in part to immunosuppressive mechanisms that allow tumor cells to evade destruction by immune cells. One of the hallmarks of immune suppression is the paucity of tumor-infiltrating lymphocytes (TILs), characterized by low numbers of effector CD4+ and CD8+ T cells in the tumor microenvironment (TME). Additionally, the proper activation and function of lymphocytes that successfully infiltrate the tumor are hampered by the lack of co-stimulatory molecules and the increase in inhibitory factors. These contribute to the imbalance of effector functions by natural killer (NK) and T cells and the immunosuppressive functions by myeloid-derived suppressor cells (MDSCs) and regulatory T cells (Tregs) in the TME, resulting in a dysfunctional anti-tumor immune response. Therefore, therapeutic regimens that elicit immune responses and reverse immune dysfunction are required to counter immune suppression in the TME and allow for the re-establishment of proper immune surveillance. Immuno-oncology (IO) agents, such as immune checkpoint blockade and TGF-β trapping molecules, have been developed to decrease or block suppressive factors to enable the activity of effector cells in the TME. Therapeutic agents that target immunosuppressive cells, either by direct lysis or altering their functions, have also been demonstrated to decrease the barrier to effective immune response. Other therapies, such as tumor antigen-specific vaccines and immunocytokines, have been shown to activate and improve the recruitment of CD4+ and CD8+ T cells to the tumor, resulting in improved T effector to Treg ratio. The preclinical data on these diverse IO agents have led to the development of ongoing phase I and II clinical trials. This review aims to provide an overview of select therapeutic strategies that tip the balance from immunosuppression to immune activity in the TME.
Collapse
|
4
|
Lao J, Cao C, Niu X, Deng S, Ming S, Liang S, Shang Y, Yuan Y, Shi X, Liang Z, Wu M, Wu Y. OX40 enhances T cell immune response to PD-1 blockade therapy in non-small cell lung cancer. Int Immunopharmacol 2022; 108:108813. [DOI: 10.1016/j.intimp.2022.108813] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2021] [Revised: 04/14/2022] [Accepted: 04/26/2022] [Indexed: 11/28/2022]
|
5
|
Mascarelli DE, Rosa RSM, Toscaro JM, Semionatto IF, Ruas LP, Fogagnolo CT, Lima GC, Bajgelman MC. Boosting Antitumor Response by Costimulatory Strategies Driven to 4-1BB and OX40 T-cell Receptors. Front Cell Dev Biol 2021; 9:692982. [PMID: 34277638 PMCID: PMC8277962 DOI: 10.3389/fcell.2021.692982] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2021] [Accepted: 05/27/2021] [Indexed: 01/01/2023] Open
Abstract
Immunotherapy explores several strategies to enhance the host immune system’s ability to detect and eliminate cancer cells. The use of antibodies that block immunological checkpoints, such as anti–programed death 1/programed death 1 ligand and cytotoxic T-lymphocyte–associated protein 4, is widely recognized to generate a long-lasting antitumor immune response in several types of cancer. Evidence indicates that the elimination of tumors by T cells is the key for tumor control. It is well known that costimulatory and coinhibitory pathways are critical regulators in the activation of T cells. Besides blocking checkpoints inhibitors, the agonistic signaling on costimulatory molecules also plays an important role in T-cell activation and antitumor response. Therefore, molecules driven to costimulatory pathways constitute promising targets in cancer therapy. The costimulation of tumor necrosis factor superfamily receptors on lymphocytes surface may transduce signals that control the survival, proliferation, differentiation, and effector functions of these immune cells. Among the members of the tumor necrosis factor receptor superfamily, there are 4-1BB and OX40. Several clinical studies have been carried out targeting these molecules, with agonist monoclonal antibodies, and preclinical studies exploring their ligands and other experimental approaches. In this review, we discuss functional aspects of 4-1BB and OX40 costimulation, as well as the progress of its application in immunotherapies.
Collapse
Affiliation(s)
- Daniele E Mascarelli
- Brazilian Biosciences National Laboratory (LNBio), Brazilian Center for Research in Energy and Materials (CNPEM), Campinas, Brazil.,Faculty of Pharmaceutical Sciences, University of Campinas, Campinas, Brazil
| | - Rhubia S M Rosa
- Brazilian Biosciences National Laboratory (LNBio), Brazilian Center for Research in Energy and Materials (CNPEM), Campinas, Brazil.,Faculty of Pharmaceutical Sciences, University of Campinas, Campinas, Brazil
| | - Jessica M Toscaro
- Brazilian Biosciences National Laboratory (LNBio), Brazilian Center for Research in Energy and Materials (CNPEM), Campinas, Brazil.,Medical School, University of Campinas (UNICAMP), Campinas, Brazil
| | - Isadora F Semionatto
- Brazilian Biosciences National Laboratory (LNBio), Brazilian Center for Research in Energy and Materials (CNPEM), Campinas, Brazil.,Faculty of Pharmaceutical Sciences, University of Campinas, Campinas, Brazil
| | - Luciana P Ruas
- Brazilian Biosciences National Laboratory (LNBio), Brazilian Center for Research in Energy and Materials (CNPEM), Campinas, Brazil
| | - Carolinne T Fogagnolo
- Brazilian Biosciences National Laboratory (LNBio), Brazilian Center for Research in Energy and Materials (CNPEM), Campinas, Brazil.,Medical School of Ribeirão Preto (FMRP), University of São Paulo, Ribeirão Preto, Brazil
| | - Gabriel C Lima
- Brazilian Biosciences National Laboratory (LNBio), Brazilian Center for Research in Energy and Materials (CNPEM), Campinas, Brazil.,Pro Rectory of Graduation, University of São Paulo, São Paulo, Brazil
| | - Marcio C Bajgelman
- Brazilian Biosciences National Laboratory (LNBio), Brazilian Center for Research in Energy and Materials (CNPEM), Campinas, Brazil.,Faculty of Pharmaceutical Sciences, University of Campinas, Campinas, Brazil.,Medical School, University of Campinas (UNICAMP), Campinas, Brazil
| |
Collapse
|
6
|
Iwasaki-Hozumi H, Chagan-Yasutan H, Ashino Y, Hattori T. Blood Levels of Galectin-9, an Immuno-Regulating Molecule, Reflect the Severity for the Acute and Chronic Infectious Diseases. Biomolecules 2021; 11:biom11030430. [PMID: 33804076 PMCID: PMC7998537 DOI: 10.3390/biom11030430] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2020] [Revised: 03/08/2021] [Accepted: 03/10/2021] [Indexed: 02/07/2023] Open
Abstract
Galectin-9 (Gal-9) is a β-galactoside-binding lectin capable of promoting or suppressing the progression of infectious diseases. This protein is susceptible to cleavage of its linker-peptides by several proteases, and the resulting cleaved forms, N-terminal carbohydrate recognition domain (CRD) and C-terminal CRD, bind to various glycans. It has been suggested that full-length (FL)-Gal-9 and the truncated (Tr)-Gal-9s could exert different functions from one another via their different glycan-binding activities. We propose that FL-Gal-9 regulates the pathogenesis of infectious diseases, including human immunodeficiency virus (HIV) infection, HIV co-infected with opportunistic infection (HIV/OI), dengue, malaria, leptospirosis, and tuberculosis (TB). We also suggest that the blood levels of FL-Gal-9 reflect the severity of dengue, malaria, and HIV/OI, and those of Tr-Gal-9 markedly reflect the severity of HIV/OI. Recently, matrix metallopeptidase-9 (MMP-9) was suggested to be an indicator of respiratory failure from coronavirus disease 2019 (COVID-19) as well as useful for differentiating pulmonary from extrapulmonary TB. The protease cleavage of FL-Gal-9 may lead to uncontrolled hyper-immune activation, including a cytokine storm. In summary, Gal-9 has potential to reflect the disease severity for the acute and chronic infectious diseases.
Collapse
Affiliation(s)
- Hiroko Iwasaki-Hozumi
- Department of Health Science and Social Welfare, Kibi International University, Takahashi 716-8508, Japan; (H.I.-H.); (H.C.-Y.)
| | - Haorile Chagan-Yasutan
- Department of Health Science and Social Welfare, Kibi International University, Takahashi 716-8508, Japan; (H.I.-H.); (H.C.-Y.)
- Mongolian Psychosomatic Medicine Department, International Mongolian Medicine Hospital of Inner Mongolia, Hohhot 010065, China
| | - Yugo Ashino
- Department of Respiratory Medicine, Sendai City Hospital, Sendai 982-8502, Japan;
| | - Toshio Hattori
- Department of Health Science and Social Welfare, Kibi International University, Takahashi 716-8508, Japan; (H.I.-H.); (H.C.-Y.)
- Correspondence: ; Tel.: +81-866-22-9454
| |
Collapse
|
7
|
Kim SI, Cassella CR, Byrne KT. Tumor Burden and Immunotherapy: Impact on Immune Infiltration and Therapeutic Outcomes. Front Immunol 2021; 11:629722. [PMID: 33597954 PMCID: PMC7882695 DOI: 10.3389/fimmu.2020.629722] [Citation(s) in RCA: 94] [Impact Index Per Article: 23.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2020] [Accepted: 12/18/2020] [Indexed: 12/20/2022] Open
Abstract
Cancer immunotherapy has revolutionized the treatment landscape in medical oncology, but its efficacy has been variable across patients. Biomarkers to predict such differential response to immunotherapy include cytotoxic T lymphocyte infiltration, tumor mutational burden, and microsatellite instability. A growing number of studies also suggest that baseline tumor burden, or tumor size, predicts response to immunotherapy. In this review, we discuss the changes in immune profile and therapeutic responses that occur with increasing tumor size. We also overview therapeutic approaches to reduce tumor burden and favorably modulate the immune microenvironment of larger tumors.
Collapse
Affiliation(s)
- Samuel I Kim
- Program in Biochemistry, College of Arts and Sciences, University of Pennsylvania, Philadelphia, PA, United States
| | - Christopher R Cassella
- Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, United States
| | - Katelyn T Byrne
- Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, United States.,Parker Institute for Cancer Immunotherapy, University of Pennsylvania, Philadelphia, PA, United States
| |
Collapse
|
8
|
Morales Del Valle C, Maxwell JR, Xu MM, Menoret A, Mittal P, Tsurutani N, Adler AJ, Vella AT. Costimulation Induces CD4 T Cell Antitumor Immunity via an Innate-like Mechanism. Cell Rep 2020; 27:1434-1445.e3. [PMID: 31042471 DOI: 10.1016/j.celrep.2019.04.016] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2018] [Revised: 12/12/2018] [Accepted: 04/02/2019] [Indexed: 12/12/2022] Open
Abstract
Chronic exposure to tumor-associated antigens inactivates cognate T cells, restricting the repertoire of tumor-specific effector T cells. This problem was studied here by transferring TCR transgenic CD4 T cells into recipient mice that constitutively express a cognate self-antigen linked to MHC II on CD11c-bearing cells. Immunotherapeutic agonists to CD134 plus CD137, "dual costimulation," induces specific CD4 T cell expansion and expression of the receptor for the Th2-associated IL-1 family cytokine IL-33. Rather than producing IL-4, however, they express the tumoricidal Th1 cytokine IFNγ when stimulated with IL-33 or IL-36 (a related IL-1 family member) plus IL-12 or IL-2. IL-36, which is induced within B16-F10 melanomas by dual costimulation, reduces tumor growth when injected intratumorally as a monotherapy and boosts the efficacy of tumor-nonspecific dual costimulated CD4 T cells. Dual costimulation thus enables chronic antigen-exposed CD4 T cells, regardless of tumor specificity, to elaborate tumoricidal function in response to tumor-associated cytokines.
Collapse
Affiliation(s)
| | - Joseph R Maxwell
- Department of Immunology, School of Medicine, UConn Health, Farmington, CT 06030, USA
| | - Maria M Xu
- Department of Immunology, School of Medicine, UConn Health, Farmington, CT 06030, USA
| | - Antoine Menoret
- Department of Immunology, School of Medicine, UConn Health, Farmington, CT 06030, USA
| | - Payal Mittal
- Department of Immunology, School of Medicine, UConn Health, Farmington, CT 06030, USA
| | - Naomi Tsurutani
- Department of Immunology, School of Medicine, UConn Health, Farmington, CT 06030, USA
| | - Adam J Adler
- Department of Immunology, School of Medicine, UConn Health, Farmington, CT 06030, USA.
| | - Anthony T Vella
- Department of Immunology, School of Medicine, UConn Health, Farmington, CT 06030, USA.
| |
Collapse
|
9
|
Roy S, Sethi TK, Taylor D, Kim YJ, Johnson DB. Breakthrough concepts in immune-oncology: Cancer vaccines at the bedside. J Leukoc Biol 2020; 108:1455-1489. [PMID: 32557857 DOI: 10.1002/jlb.5bt0420-585rr] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2019] [Revised: 04/15/2020] [Accepted: 04/18/2020] [Indexed: 12/11/2022] Open
Abstract
Clinical approval of the immune checkpoint blockade (ICB) agents for multiple cancer types has reinvigorated the long-standing work on cancer vaccines. In the pre-ICB era, clinical efforts focused on the Ag, the adjuvants, the formulation, and the mode of delivery. These translational efforts on therapeutic vaccines range from cell-based (e.g., dendritic cells vaccine Sipuleucel-T) to DNA/RNA-based platforms with various formulations (liposome), vectors (Listeria monocytogenes), or modes of delivery (intratumoral, gene gun, etc.). Despite promising preclinical results, cancer vaccine trials without ICB have historically shown little clinical activity. With the anticipation and expansion of combinatorial immunotherapeutic trials with ICB, the cancer vaccine field has entered the personalized medicine arena with recent advances in immunogenic neoantigen-based vaccines. In this article, we review the literature to organize the different cancer vaccines in the clinical space, and we will discuss their advantages, limits, and recent progress to overcome their challenges. Furthermore, we will also discuss recent preclinical advances and clinical strategies to combine vaccines with checkpoint blockade to improve therapeutic outcome and present a translational perspective on future directions.
Collapse
Affiliation(s)
- Sohini Roy
- Department of Otolaryngology - Head & Neck Surgery, Vanderbilt University Medical Center, Nashville, Tennessee, USA
| | - Tarsheen K Sethi
- Department of Medicine, Vanderbilt University Medical Center, Nashville, Tennessee, USA
| | - David Taylor
- Department of Otolaryngology - Head & Neck Surgery, Vanderbilt University Medical Center, Nashville, Tennessee, USA
| | - Young J Kim
- Department of Otolaryngology - Head & Neck Surgery, Vanderbilt University Medical Center, Nashville, Tennessee, USA
| | - Douglas B Johnson
- Department of Medicine, Vanderbilt University Medical Center, Nashville, Tennessee, USA
| |
Collapse
|
10
|
Fu Y, Lin Q, Zhang Z, Zhang L. Therapeutic strategies for the costimulatory molecule OX40 in T-cell-mediated immunity. Acta Pharm Sin B 2020; 10:414-433. [PMID: 32140389 PMCID: PMC7049610 DOI: 10.1016/j.apsb.2019.08.010] [Citation(s) in RCA: 170] [Impact Index Per Article: 34.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2019] [Revised: 07/18/2019] [Accepted: 07/19/2019] [Indexed: 12/11/2022] Open
Abstract
The T cell co-stimulatory molecule OX40 and its cognate ligand OX40L have attracted broad research interest as a therapeutic target in T cell-mediated diseases. Accumulating preclinical evidence highlights the therapeutic efficacy of both agonist and blockade of the OX40-OX40L interaction. Despite this progress, many questions about the immuno-modulator roles of OX40 on T cell function remain unanswered. In this review we summarize the impact of the OX40-OX40L interaction on T cell subsets, including Th1, Th2, Th9, Th17, Th22, Treg, Tfh, and CD8+ T cells, to gain a comprehensive understanding of anti-OX40 mAb-based therapies. The potential therapeutic application of the OX40-OX40L interaction in autoimmunity diseases and cancer immunotherapy are further discussed; OX40-OX40L blockade may ameliorate autoantigen-specific T cell responses and reduce immune activity in autoimmunity diseases. We also explore the rationale of targeting OX40-OX40L interactions in cancer immunotherapy. Ligation of OX40 with targeted agonist anti-OX40 mAbs conveys activating signals to T cells. When combined with other therapeutic treatments, such as anti-PD-1 or anti-CTLA-4 blockade, cytokines, chemotherapy, or radiotherapy, the anti-tumor activity of agonist anti-OX40 treatment will be further enhanced. These data collectively suggest great potential for OX40-mediated therapies.
Collapse
Affiliation(s)
- Yu Fu
- Key Laboratory of Drug Targeting and Drug Delivery System of the Education Ministry, Sichuan Engineering Laboratory for Plant-Sourced Drug and Sichuan Research Center for Drug Precision Industrial Technology, College of Polymer Science and Engineering, West China School of Pharmacy, Sichuan University, Chengdu 610064, China
- Department of Pharmacology, Yale University School of Medicine, New Haven, CT 06510, USA
| | - Qing Lin
- Key Laboratory of Drug Targeting and Drug Delivery System of the Education Ministry, Sichuan Engineering Laboratory for Plant-Sourced Drug and Sichuan Research Center for Drug Precision Industrial Technology, College of Polymer Science and Engineering, West China School of Pharmacy, Sichuan University, Chengdu 610064, China
| | - Zhirong Zhang
- Key Laboratory of Drug Targeting and Drug Delivery System of the Education Ministry, Sichuan Engineering Laboratory for Plant-Sourced Drug and Sichuan Research Center for Drug Precision Industrial Technology, College of Polymer Science and Engineering, West China School of Pharmacy, Sichuan University, Chengdu 610064, China
| | - Ling Zhang
- Key Laboratory of Drug Targeting and Drug Delivery System of the Education Ministry, Sichuan Engineering Laboratory for Plant-Sourced Drug and Sichuan Research Center for Drug Precision Industrial Technology, College of Polymer Science and Engineering, West China School of Pharmacy, Sichuan University, Chengdu 610064, China
| |
Collapse
|
11
|
Clearance of Chikungunya Virus Infection in Lymphoid Tissues Is Promoted by Treatment with an Agonistic Anti-CD137 Antibody. J Virol 2019; 93:JVI.01231-19. [PMID: 31578287 DOI: 10.1128/jvi.01231-19] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2019] [Accepted: 09/19/2019] [Indexed: 01/13/2023] Open
Abstract
CD137, a member of the tumor necrosis factor receptor superfamily of cell surface proteins, acts as a costimulatory receptor on T cells, natural killer cells, B cell subsets, and some dendritic cells. Agonistic anti-CD137 monoclonal antibody (MAb) therapy has been combined with other chemotherapeutic agents in human cancer trials. Based on its ability to promote tumor clearance, we hypothesized that anti-CD137 MAb might activate immune responses and resolve chronic viral infections. We evaluated anti-CD137 MAb therapy in a mouse infection model of chikungunya virus (CHIKV), an alphavirus that causes chronic polyarthritis in humans and is associated with reservoirs of CHIKV RNA that are not cleared efficiently by adaptive immune responses. Analysis of viral tropism revealed that CHIKV RNA was present preferentially in splenic B cells and follicular dendritic cells during the persistent phase of infection, and animals lacking B cells did not develop persistent CHIKV infection in lymphoid tissue. Anti-CD137 MAb treatment resulted in T cell-dependent clearance of CHIKV RNA in lymphoid tissue, although this effect was not observed in musculoskeletal tissue. The clearance of CHIKV RNA from lymphoid tissue by anti-CD137 MAb was associated with reductions in the numbers of germinal center B cells and follicular dendritic cells. Similar results were observed with anti-CD137 MAb treatment of mice infected with Mayaro virus, a related arthritogenic alphavirus. Thus, anti-CD137 MAb treatment promotes resolution of chronic alphavirus infection in lymphoid tissues by reducing the numbers of target cells for infection and persistence.IMPORTANCE Although CHIKV causes persistent infection in lymphoid and musculoskeletal tissues in multiple animals, the basis for this is poorly understood, which has hampered pharmacological efforts to promote viral clearance. Here, we evaluated the therapeutic effects on persistent CHIKV infection of an agonistic anti-CD137 MAb that can activate T cell and natural killer cell responses to clear tumors. We show that treatment with anti-CD137 MAb promotes the clearance of persistent alphavirus RNA from lymphoid but not musculoskeletal tissues. This occurs because anti-CD137 MAb-triggered T cells reduce the numbers of target germinal center B cells and follicular dendritic cells, which are the primary reservoirs for CHIKV in the spleen and lymph nodes. Our studies help to elucidate the basis for CHIKV persistence and begin to provide strategies that can clear long-term cellular reservoirs of infection.
Collapse
|
12
|
Konstorum A, Vella AT, Adler AJ, Laubenbacher RC. A mathematical model of combined CD8 T cell costimulation by 4-1BB (CD137) and OX40 (CD134) receptors. Sci Rep 2019; 9:10862. [PMID: 31350431 PMCID: PMC6659676 DOI: 10.1038/s41598-019-47333-y] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2018] [Accepted: 07/11/2019] [Indexed: 02/07/2023] Open
Abstract
Combined agonist stimulation of the TNFR costimulatory receptors 4-1BB (CD137) and OX40(CD134) has been shown to generate supereffector CD8 T cells that clonally expand to greater levels, survive longer, and produce a greater quantity of cytokines compared to T cells stimulated with an agonist of either costimulatory receptor individually. In order to understand the mechanisms for this effect, we have created a mathematical model for the activation of the CD8 T cell intracellular signaling network by mono- or dual-costimulation. We show that supereffector status is generated via downstream interacting pathways that are activated upon engagement of both receptors, and in silico simulations of the model are supported by published experimental results. The model can thus be used to identify critical molecular targets of T cell dual-costimulation in the context of cancer immunotherapy.
Collapse
Affiliation(s)
- Anna Konstorum
- Center for Quantitative Medicine, School of Medicine, UConn Health, 263 Farmington Ave., Farmington, CT, USA.
| | - Anthony T Vella
- Department of Immunology, School of Medicine, UConn Health, 263 Farmington Ave., Farmington, CT, USA
| | - Adam J Adler
- Department of Immunology, School of Medicine, UConn Health, 263 Farmington Ave., Farmington, CT, USA
| | - Reinhard C Laubenbacher
- Center for Quantitative Medicine, School of Medicine, UConn Health, 263 Farmington Ave., Farmington, CT, USA.,Jackson Laboratory for Genomic Medicine, 263 Farmington Ave., Farmington, CT, USA
| |
Collapse
|
13
|
Vermaelen K. Vaccine Strategies to Improve Anti-cancer Cellular Immune Responses. Front Immunol 2019; 10:8. [PMID: 30723469 PMCID: PMC6349827 DOI: 10.3389/fimmu.2019.00008] [Citation(s) in RCA: 142] [Impact Index Per Article: 23.7] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2018] [Accepted: 01/03/2019] [Indexed: 12/24/2022] Open
Abstract
More than many other fields in medicine, cancer vaccine development has been plagued by a wide gap between the massive amounts of highly encouraging preclinical data on one hand, and the disappointing clinical results on the other. It is clear now that traditional approaches from the infectious diseases' vaccine field cannot be borrowed as such to treat cancer. This review highlights some of the strategies developed to improve vaccine formulations for oncology, including research into more powerful or “smarter” adjuvants to elicit anti-tumoral cellular immune responses. As an illustration of the difficulties in translating smart preclinical strategies into real benefit for the cancer patient, the difficult road of vaccine development in lung cancer is given as example. Finally, an outline is provided of the combinatorial strategies that leverage the increasing knowledge on tumor-associated immune suppressive networks. Indeed, combining with drugs that target the dominant immunosuppressive pathway in a given tumor promises to unlock the true power of cancer vaccines and potentially offer long-term protection from disease relapse.
Collapse
Affiliation(s)
- Karim Vermaelen
- Tumor Immunology Laboratory, Department of Pulmonary Medicine and Immuno-Oncology Network Ghent, Ghent University Hospital, Ghent, Belgium
| |
Collapse
|
14
|
Nanni P, De Giovanni C, Burocchi A, Nicoletti G, Landuzzi L, Palladini A, Ianzano ML, Arioli I, Colombo MP, Lollini PL. OX40 triggering concomitant to IL12-engineered cell vaccine hampers the immunoprevention of HER2/neu-driven mammary carcinogenesis. Oncoimmunology 2018; 7:e1465164. [PMID: 30221061 PMCID: PMC6136865 DOI: 10.1080/2162402x.2018.1465164] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2018] [Revised: 04/09/2018] [Accepted: 04/09/2018] [Indexed: 10/24/2022] Open
Abstract
This study evaluated the effects of combining an OX40 agonistic antibody (aOX40) with a cell vaccine targeting HER2/neu, called "Triplex". Such HER2/neu cell vaccine included two biological adjuvants (interleukin 12 (IL12) and allogeneic histocompatibility antigens) and was previously found able to prevent autochthonous HER2/neu-driven mammary carcinogenesis. Timing of aOX40 administration, concomitantly or after cell vaccination, gave opposite results. Unexpectedly, vaccine efficacy was hampered by concomitant OX40 triggering. Such decreased immunoprevention was likely due to a reduced induction of anti-HER2/neu antibodies and to a higher level of Treg activation. On the contrary, aOX40 administration after the completion of vaccination slightly but significantly increased immunopreventive vaccine efficacy, and led to increased production of GM-CSF and IL10. In conclusion, OX40 triggering can either impair or ameliorate immunoprevention of HER2/neu-driven mammary carcinogenesis depending on the schedule of aOX40 administration.
Collapse
Affiliation(s)
- Patrizia Nanni
- Laboratory of Immunology and Biology of Metastasis, Department of Experimental, Diagnostic and Specialty Medicine, University of Bologna, Italy
| | - Carla De Giovanni
- Laboratory of Immunology and Biology of Metastasis, Department of Experimental, Diagnostic and Specialty Medicine, University of Bologna, Italy
| | - Alessia Burocchi
- Molecular Immunology Unit, Department of Experimental Oncology and Molecular Medicine, Fondazione IRCCS Istituto Nazionale dei Tumori, Milano, Italy
| | - Giordano Nicoletti
- Rizzoli Orthopedic Institute, Laboratory of Experimental Oncology, Bologna, Italy
| | - Lorena Landuzzi
- Rizzoli Orthopedic Institute, Laboratory of Experimental Oncology, Bologna, Italy
| | - Arianna Palladini
- Laboratory of Immunology and Biology of Metastasis, Department of Experimental, Diagnostic and Specialty Medicine, University of Bologna, Italy
| | - Marianna Lucia Ianzano
- Laboratory of Immunology and Biology of Metastasis, Department of Experimental, Diagnostic and Specialty Medicine, University of Bologna, Italy
| | - Ivano Arioli
- Molecular Immunology Unit, Department of Experimental Oncology and Molecular Medicine, Fondazione IRCCS Istituto Nazionale dei Tumori, Milano, Italy
| | - Mario P Colombo
- Molecular Immunology Unit, Department of Experimental Oncology and Molecular Medicine, Fondazione IRCCS Istituto Nazionale dei Tumori, Milano, Italy
| | - Pier-Luigi Lollini
- Laboratory of Immunology and Biology of Metastasis, Department of Experimental, Diagnostic and Specialty Medicine, University of Bologna, Italy
| |
Collapse
|
15
|
A novel biologic platform elicits profound T cell costimulatory activity and antitumor immunity in mice. Cancer Immunol Immunother 2018; 67:605-613. [PMID: 29327109 DOI: 10.1007/s00262-018-2116-1] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2017] [Accepted: 01/03/2018] [Indexed: 02/06/2023]
Abstract
Combination immunotherapies utilizing complementary modalities that target distinct tumor attributes or immunosuppressive mechanisms, or engage different arms of the antitumor immune response, can elicit greater therapeutic efficacy than the component monotherapies. Increasing the number of agents included in a therapeutic cocktail can further increase efficacy, however, this approach poses numerous challenges for clinical translation. Here, a novel platform to simplify combination immunotherapy by covalently linking immunotherapeutic agonists to the costimulatory receptors CD134 and CD137 into a single heterodimeric drug, "OrthomAb", is shown. This reagent not only retains costimulatory T cell activity, but also elicits unique T cell functions that are not programmed by either individual agonist, and preferentially expands effector T cells over Tregs. Finally, in an aggressive melanoma model OrthomAb elicits better therapeutic efficacy compared to the unlinked agonists. This demonstration that two drugs can be combined into one provides a framework for distilling complex combination drug cocktails into simpler delivery platforms.
Collapse
|
16
|
Mittal P, Abblett R, Ryan JM, Hagymasi AT, Agyekum-Yamoah A, Svedova J, Reiner SL, St Rose MC, Hanley MP, Vella AT, Adler AJ. An Immunotherapeutic CD137 Agonist Releases Eomesodermin from ThPOK Repression in CD4 T Cells. THE JOURNAL OF IMMUNOLOGY 2018; 200:1513-1526. [PMID: 29305435 DOI: 10.4049/jimmunol.1701039] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/18/2017] [Accepted: 12/06/2017] [Indexed: 12/24/2022]
Abstract
Agonists to the TNF/TNFR costimulatory receptors CD134 (OX40) and CD137 (4-1BB) elicit antitumor immunity. Dual costimulation with anti-CD134 plus anti-CD137 is particularly potent because it programs cytotoxic potential in CD8+ and CD4+ T cells. Cytotoxicity in dual-costimulated CD4 T cells depends on the T-box transcription factor eomesodermin (Eomes), which we report is induced via a mechanism that does not rely on IL-2, in contrast to CD8+ CTL, but rather depends on the CD8 T cell lineage commitment transcription factor Runx3, which supports Eomes expression in mature CD8+ CTLs. Further, Eomes and Runx3 were indispensable for dual-costimulated CD4 T cells to mediate antitumor activity in an aggressive melanoma model. Runx3 is also known to be expressed in standard CD4 Th1 cells where it fosters IFN-γ expression; however, the CD4 T cell lineage commitment factor ThPOK represses transcription of Eomes and other CD8 lineage genes, such as Cd8a Hence, CD4 T cells can differentiate into Eomes+ cytotoxic CD4+CD8+ double-positive T cells by terminating ThPOK expression. In contrast, dual-costimulated CD4 T cells express Eomes, despite the continued expression of ThPOK and the absence of CD8α, indicating that Eomes is selectively released from ThPOK repression. Finally, although Eomes was induced by CD137 agonist, but not CD134 agonist, administered individually, CD137 agonist failed to induce CD134-/- CD4 T cells to express Eomes or Runx3, indicating that both costimulatory pathways are required for cytotoxic Th1 programming, even when only CD137 is intentionally engaged with a therapeutic agonist.
Collapse
Affiliation(s)
- Payal Mittal
- Department of Immunology, University of Connecticut School of Medicine, Farmington, CT 06030
| | - Rebecca Abblett
- Department of Immunology, University of Connecticut School of Medicine, Farmington, CT 06030
| | - Joseph M Ryan
- Department of Immunology, University of Connecticut School of Medicine, Farmington, CT 06030
| | - Adam T Hagymasi
- Department of Immunology, University of Connecticut School of Medicine, Farmington, CT 06030
| | | | - Julia Svedova
- Department of Immunology, University of Connecticut School of Medicine, Farmington, CT 06030
| | - Steven L Reiner
- Department of Microbiology and Immunology, College of Physicians and Surgeons, Columbia University, New York, NY 10032; and.,Department of Pediatrics, College of Physicians and Surgeons, Columbia University, New York, NY 10032
| | - Marie-Clare St Rose
- Department of Immunology, University of Connecticut School of Medicine, Farmington, CT 06030
| | - Matthew P Hanley
- Department of Immunology, University of Connecticut School of Medicine, Farmington, CT 06030
| | - Anthony T Vella
- Department of Immunology, University of Connecticut School of Medicine, Farmington, CT 06030
| | - Adam J Adler
- Department of Immunology, University of Connecticut School of Medicine, Farmington, CT 06030;
| |
Collapse
|
17
|
Sapski S, Beha N, Kontermann R, Müller D. Tumor-targeted costimulation with antibody-fusion proteins improves bispecific antibody-mediated immune response in presence of immunosuppressive factors. Oncoimmunology 2017; 6:e1361594. [PMID: 29209565 DOI: 10.1080/2162402x.2017.1361594] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2017] [Revised: 07/25/2017] [Accepted: 07/26/2017] [Indexed: 10/19/2022] Open
Abstract
Therapeutic strategies aiming for the induction of an effective immune response at the tumor site can be severely hampered by the encounter of an immunosuppressive microenvironment. We investigated here the potential of concerted costimulation by tumor-directed antibody-fusion proteins with B7.1, 4-1BBL and OX40L to enforce bispecific antibody-induced T cell stimulation in presence of recognized immunosuppressive factors including IL-10, TGF-β, indoleamine 2,3-dioxygenase (IDO), PD-L1 and regulatory T cells. The expression and activity of these factors was demonstrated in the HT1080-FAP/PBMC co-culture setting, where individual and combined costimulation were still capable to enhance T cell stimulation, even though the general activation level was reduced. Additional blockade of TGF-ß or PD-1 resulted especially effective in further enhancing the degree of T cell activation. Here, best outcome was achieved by combined costimulation of targeted 4-1BBL and B7.1. Furthermore, their individual impact on the proliferation of naïve, memory and effector CD8+ and CD4+ T cell subsets, suggest the coverage of a comprehensive T cell response. Thus, our costimulatory antibody-fusion proteins show great potential to support T cell activation in adverse conditions dictated by the tumor microenvironment.
Collapse
Affiliation(s)
- Sabrina Sapski
- Institute of Cell Biology and Immunology, University of Stuttgart, Allmandring 31, Stuttgart, Germany
| | - Nadine Beha
- Institute of Cell Biology and Immunology, University of Stuttgart, Allmandring 31, Stuttgart, Germany
| | - Roland Kontermann
- Institute of Cell Biology and Immunology, University of Stuttgart, Allmandring 31, Stuttgart, Germany
| | - Dafne Müller
- Institute of Cell Biology and Immunology, University of Stuttgart, Allmandring 31, Stuttgart, Germany
| |
Collapse
|
18
|
Malamas AS, Hammond SA, Schlom J, Hodge JW. Combination therapy with an OX40L fusion protein and a vaccine targeting the transcription factor twist inhibits metastasis in a murine model of breast cancer. Oncotarget 2017; 8:90825-90841. [PMID: 29207606 PMCID: PMC5710887 DOI: 10.18632/oncotarget.19967] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2017] [Accepted: 07/23/2017] [Indexed: 12/22/2022] Open
Abstract
OX40 is a costimulatory receptor that potentiates proliferation, survival, memory formation, and effector function of CD4+ and CD8+ T-cells, while overcoming the suppressive activity of regulatory T-cells (Tregs). Here, we explored the combination of an OX40L fusion protein (OX40L-FP) with a poxvirus-based cancer vaccine (MVA-Twist-TRICOM) to inhibit tumor metastasis in the 4T1 murine breast cancer model. Contrary to the single agent treatments, the combination therapy significantly decreased the number of metastatic colonies per lung and prolonged survival. Depletion studies demonstrated that these effects were mediated by both CD4+ and CD8+ T-cells. The combination therapy a) increased the total number of T-cells in the CD4+Foxp3- population and the CD4+ central and effector memory subsets within the lung, spleen, and draining lymph node, b) enhanced infiltration of CD4+ T-cells into metastatic areas of the lung, and (c) increased the number of functional CD8+ T-cells that produced IFNγ and TNFα. The combination therapy also promoted the development of KLRG1-CD127+ memory precursor CD8+ T-cells, while reducing those with a KLRG1+ terminally differentiated phenotype. Moreover, the combination of OX40L-FP and vaccine induced greater CD4+ and CD8+ Twist-specific responses. In addition, Tregs isolated from mice receiving the combination were also less immunosuppressive in ex-vivo proliferation assays than those from the OX40L-FP and MVA-Twist-TRICOM monotherapy groups. Such results provide the rationale to combine co-stimulatory agonists with cancer vaccines for the treatment of tumor metastasis.
Collapse
Affiliation(s)
- Anthony S Malamas
- Laboratory of Tumor Immunology and Biology, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, Maryland, USA
| | | | - Jeffrey Schlom
- Laboratory of Tumor Immunology and Biology, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, Maryland, USA
| | - James W Hodge
- Laboratory of Tumor Immunology and Biology, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, Maryland, USA
| |
Collapse
|
19
|
Adler AJ, Mittal P, Ryan JM, Zhou B, Wasser JS, Vella AT. Cytokines and metabolic factors regulate tumoricidal T-cell function during cancer immunotherapy. Immunotherapy 2017; 9:71-82. [PMID: 28000531 DOI: 10.2217/imt-2016-0097] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
Recent advances in cancer biology and genetics have fostered precision therapies targeting tumor-specific attributes. Immune-based therapies that elicit cytolytic T cells (CTL) specific for tumor antigens can provide therapeutic benefit to cancer patients, however, cure rates are typically low. This largely results from immunosuppressive mechanisms operating within the tumor microenvironment, many of which inflict metabolic stresses upon CTL. Conversely, immunotherapies can mitigate specific metabolic stressors. For instance, dual costimulation immunotherapy with CD134 (OX40) plus CD137 (4-1BB) agonists appears to mediate tumor control in part by engaging cytokine networks that enable infiltrating CTL to compete for limiting supplies of glucose. Future efforts combining modalities that endow CTL with complimentary metabolic advantages should improve therapeutic efficacies.
Collapse
Affiliation(s)
- Adam J Adler
- Department of Immunology, University of Connecticut School of Medicine, Farmington, CT 06030, USA.,Department of Medicine, University of Connecticut School of Medicine, Farmington, CT 06030, USA
| | - Payal Mittal
- Department of Immunology, University of Connecticut School of Medicine, Farmington, CT 06030, USA
| | - Joseph M Ryan
- Department of Immunology, University of Connecticut School of Medicine, Farmington, CT 06030, USA
| | - Beiyan Zhou
- Department of Immunology, University of Connecticut School of Medicine, Farmington, CT 06030, USA
| | - Jeffrey S Wasser
- Department of Immunology, University of Connecticut School of Medicine, Farmington, CT 06030, USA
| | - Anthony T Vella
- Department of Immunology, University of Connecticut School of Medicine, Farmington, CT 06030, USA
| |
Collapse
|
20
|
Economopoulou P, Kotsantis I, Psyrri A. The promise of immunotherapy in head and neck squamous cell carcinoma: combinatorial immunotherapy approaches. ESMO Open 2017; 1:e000122. [PMID: 28848660 PMCID: PMC5548974 DOI: 10.1136/esmoopen-2016-000122] [Citation(s) in RCA: 42] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2016] [Revised: 12/21/2016] [Accepted: 11/23/2016] [Indexed: 12/13/2022] Open
Abstract
The immune system plays a fundamental role in preventing cancer development by recognising and eliminating tumour cells. The recent success in the field of immunotherapy has confirmed the potential to exploit the immune response as a cancer treatment. Head and neck squamous cell carcinoma (HNSCC) is a malignancy characterized by dismal prognosis and high mortality rate; low survival outcomes in combination with significant toxicity of current treatment strategies highlight the necessity for novel therapeutic modalities. HNSCC is a favourable disease for immunotherapy, as immune escape plays a key role in tumour initiation and progression. T-cell checkpoint inhibitors targeting programmed cell death protein-1 have emerged as novel immunotherapy agents showing remarkable efficacy in HNSCC. However, only a minority of patients derive benefit for single-agent immunotherapies. In this regard, combinatorial immunotherapy approaches represent an alternative strategy that might increase the number of patients who respond to immunotherapy. Focusing on HNSCC, this review will summarise novel combinations of immune checkpoint blockade with other immunotherapy treatment modalities.
Collapse
Affiliation(s)
- Panagiota Economopoulou
- 2nd Department of Internal Medicine, Section of Medical Oncology, Attikon University Hospital, National and Kapodistrian University of Athens, Athens, Greece
| | - Ioannis Kotsantis
- 2nd Department of Internal Medicine, Section of Medical Oncology, Attikon University Hospital, National and Kapodistrian University of Athens, Athens, Greece
| | - Amanda Psyrri
- 2nd Department of Internal Medicine, Section of Medical Oncology, Attikon University Hospital, National and Kapodistrian University of Athens, Athens, Greece.
| |
Collapse
|
21
|
Beyranvand Nejad E, Welters MJP, Arens R, van der Burg SH. The importance of correctly timing cancer immunotherapy. Expert Opin Biol Ther 2016; 17:87-103. [PMID: 27802061 DOI: 10.1080/14712598.2017.1256388] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
INTRODUCTION The treatment options for cancer-surgery, radiotherapy and chemotherapy-are now supplemented with immunotherapy. Previously underappreciated but now gaining strong interest are the immune modulatory properties of the three conventional modalities. Moreover, there is a better understanding of the needs and potential of the different immune therapeutic platforms. Key to improved treatment will be the combinations of modalities that complete each other's shortcomings. Area covered: Tumor-specific T-cells are required for optimal immunotherapy. In this review, the authors focus on the correct timing of different types of chemotherapeutic agents or immune modulators and immunotherapeutic drugs, not only for the activation and expansion of tumor-specific T-cells but also to support and enhance their anti-tumor efficacy. Expert opinion: At an early phase of disease, clinical success can be obtained using single treatment modalities but at later disease stages, combinations of several modalities are required. The gain in success is determined by a thorough understanding of the direct and indirect immune effects of the modalities used. Profound knowledge of these effects requires optimal tuning of immunomonitoring. This will guide the appropriate combination of treatments and allow for correct sequencing the order and interval of the different therapeutic modalities.
Collapse
Affiliation(s)
- Elham Beyranvand Nejad
- a Department of Medical Oncology , Leiden University Medical Center , Leiden , The Netherlands.,b Department of Immunohematology and Blood Transfusion , Leiden University Medical Center , Leiden , The Netherlands
| | - Marij J P Welters
- a Department of Medical Oncology , Leiden University Medical Center , Leiden , The Netherlands
| | - Ramon Arens
- b Department of Immunohematology and Blood Transfusion , Leiden University Medical Center , Leiden , The Netherlands
| | - Sjoerd H van der Burg
- a Department of Medical Oncology , Leiden University Medical Center , Leiden , The Netherlands
| |
Collapse
|
22
|
Lee SH, Danishmalik SN, Sin JI. DNA vaccines, electroporation and their applications in cancer treatment. Hum Vaccin Immunother 2016; 11:1889-900. [PMID: 25984993 DOI: 10.1080/21645515.2015.1035502] [Citation(s) in RCA: 54] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
Numerous animal studies and recent clinical studies have shown that electroporation-delivered DNA vaccines can elicit robust Ag-specific CTL responses and reduce disease severity. However, cancer antigens are generally poorly immunogenic, requiring special conditions for immune response induction. To date, many different approaches have been used to elicit Ag-specific CTL and anti-neoplastic responses to DNA vaccines against cancer. In vivo electroporation is one example, whereas others include DNA manipulation, xenogeneic antigen use, immune stimulatory molecule and immune response regulator application, DNA prime-boost immunization strategy use and different DNA delivery methods. These strategies likely increase the immunogenicity of cancer DNA vaccines, thereby contributing to cancer eradication. However, cancer cells are heterogeneous and might become CTL-resistant. Thus, understanding the CTL resistance mechanism(s) employed by cancer cells is critical to develop counter-measures for this immune escape. In this review, the use of electroporation as a DNA delivery method, the strategies used to enhance the immune responses, the cancer antigens that have been tested, and the escape mechanism(s) used by tumor cells are discussed, with a focus on the progress of clinical trials using cancer DNA vaccines.
Collapse
Key Words
- AFP, α-fetoprotein
- APCs, antigen presenting cells
- CEA, carcinoembryonic antigen
- CTLA-4, cytotoxic T lymphocyte-associated antigen-4
- DCs, dendritic cells
- DNA vaccine
- EP, electroporation
- GITR, glucocorticoid-induced tumor necrosis factor receptor family-related gene
- HPV, human papillomavirus
- HSP, heat shock protein
- HSV, herpes simplex virus
- ID, intradermal
- IM, intramuscular
- MAGE, melanoma-associated antigen
- MART, melanoma antigen recognized by T cells
- PAP, prostatic acid phosphatase
- PD, programmed death
- PRAME, preferentially expressed antigen in melanoma
- PSA, prostate-specific antigen
- PSMA, prostate-specific membrane antigen
- WT1, Wilm's tumor
- anti-tumor immunity
- cancer
- hTERT, human telomerase reverse transcriptase
- tumor immune evasion
Collapse
Affiliation(s)
- Si-Hyeong Lee
- a BK21 Plus Graduate Program; Department of Microbiology ; School of Medicine; Kangwon National University ; Chuncheon , Gangwon-do , Korea
| | | | | |
Collapse
|
23
|
Ryan JM, Wasser JS, Adler AJ, Vella AT. Enhancing the safety of antibody-based immunomodulatory cancer therapy without compromising therapeutic benefit: Can we have our cake and eat it too? Expert Opin Biol Ther 2016; 16:655-74. [PMID: 26855028 DOI: 10.1517/14712598.2016.1152256] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
INTRODUCTION Monoclonal antibodies (mAbs) targeting checkpoint inhibitors have demonstrated clinical benefit in treating patients with cancer and have paved the way for additional immune-modulating mAbs such as those targeting costimulatory receptors. The full clinical utility of these agents, however, is hampered by immune-related adverse events (irAEs) that can occur during therapy. AREAS COVERED We first provide a general overview of tumor immunity, followed by a review of the two major classes of immunomodulatory mAbs being developed as cancer therapeutics: checkpoint inhibitors and costimulatory receptor agonists. We then discuss therapy-associated adverse events. Finally, we describe in detail the mechanisms driving their therapeutic activity, with an emphasis on interactions between antibody fragment crystallizable (Fc) domains and Fc receptors (FcR). EXPERT OPINION Given that Fc-FcR interactions appear critical in facilitating the ability of immunomodulatory mAbs to elicit both therapeutically useful as well as adverse effects, the engineering of mAbs that can effectively engage their targets while limiting interaction with FcRs might represent a promising future avenue for developing the next generation of immune-enhancing tumoricidal agents with increased safety and retention of efficacy.
Collapse
Affiliation(s)
- Joseph M Ryan
- a Department of Immunology , UConn Health , Farmington , CT , USA
| | | | - Adam J Adler
- a Department of Immunology , UConn Health , Farmington , CT , USA
| | - Anthony T Vella
- a Department of Immunology , UConn Health , Farmington , CT , USA
| |
Collapse
|
24
|
Sanchez-Paulete AR, Labiano S, Rodriguez-Ruiz ME, Azpilikueta A, Etxeberria I, Bolaños E, Lang V, Rodriguez M, Aznar MA, Jure-Kunkel M, Melero I. Deciphering CD137 (4-1BB) signaling in T-cell costimulation for translation into successful cancer immunotherapy. Eur J Immunol 2016; 46:513-22. [PMID: 26773716 DOI: 10.1002/eji.201445388] [Citation(s) in RCA: 99] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2015] [Revised: 12/29/2015] [Accepted: 01/11/2016] [Indexed: 01/22/2023]
Abstract
CD137 (4-1BB, TNF-receptor superfamily 9) is a surface glycoprotein of the TNFR family which can be induced on a variety of leukocyte subsets. On T and NK cells, CD137 is expressed following activation and, if ligated by its natural ligand (CD137L), conveys polyubiquitination-mediated signals via TNF receptor associated factor 2 that inhibit apoptosis, while enhancing proliferation and effector functions. CD137 thus behaves as a bona fide inducible costimulatory molecule. These functional properties of CD137 can be exploited in cancer immunotherapy by systemic administration of agonist monoclonal antibodies, which increase anticancer CTLs and enhance NK-cell-mediated antibody-dependent cell-mediated cytotoxicity. Reportedly, anti-CD137 mAb and adoptive T-cell therapy strongly synergize, since (i) CD137 expression can be used to select the T cells endowed with the best activities against the tumor, (ii) costimulation of the lymphocyte cultures to be used in adoptive T-cell therapy can be done with CD137 agonist antibodies or CD137L, and (iii) synergistic effects upon coadministration of T cells and antibodies are readily observed in mouse models. Furthermore, the signaling cytoplasmic tail of CD137 is a key component of anti-CD19 chimeric antigen receptors that are used to redirect T cells against leukemia and lymphoma in the clinic. Ongoing phase II clinical trials with agonist antibodies and the presence of CD137 sequence in these successful chimeric antigen receptors highlight the importance of CD137 in oncoimmunology.
Collapse
Affiliation(s)
- Alfonso R Sanchez-Paulete
- Division of Immunology and Immunotherapy, Center for Applied Medical Research (CIMA), University of Navarra, Pamplona, Spain.,Instituto de Investigación Sanitaria de Navarra (IdISNA), Pamplona, Spain
| | - Sara Labiano
- Division of Immunology and Immunotherapy, Center for Applied Medical Research (CIMA), University of Navarra, Pamplona, Spain.,Instituto de Investigación Sanitaria de Navarra (IdISNA), Pamplona, Spain
| | - Maria E Rodriguez-Ruiz
- Division of Immunology and Immunotherapy, Center for Applied Medical Research (CIMA), University of Navarra, Pamplona, Spain.,Instituto de Investigación Sanitaria de Navarra (IdISNA), Pamplona, Spain.,University Clinic, University of Navarra, Pamplona, Spain
| | - Arantza Azpilikueta
- Division of Immunology and Immunotherapy, Center for Applied Medical Research (CIMA), University of Navarra, Pamplona, Spain.,Instituto de Investigación Sanitaria de Navarra (IdISNA), Pamplona, Spain
| | - Iñaki Etxeberria
- Division of Immunology and Immunotherapy, Center for Applied Medical Research (CIMA), University of Navarra, Pamplona, Spain.,Instituto de Investigación Sanitaria de Navarra (IdISNA), Pamplona, Spain
| | - Elixabet Bolaños
- Division of Immunology and Immunotherapy, Center for Applied Medical Research (CIMA), University of Navarra, Pamplona, Spain.,Instituto de Investigación Sanitaria de Navarra (IdISNA), Pamplona, Spain
| | - Valérie Lang
- Ubiquitylation and Cancer Molecular Biology Laboratory, Foundation for Stem Cell Research, Fundación Inbiomed, San Sebastián, Spain
| | - Manuel Rodriguez
- Advanced Technology Institute in Life Sciences (ITAV), CNRS-USR3505, Toulouse, France.,University of Toulouse III-Paul Sabatier, Toulouse, France.,Institut de Pharmacologie et de Biologie Structurale (IPBS), CNRS-UMR5089, Toulouse, France
| | - M Angela Aznar
- Division of Immunology and Immunotherapy, Center for Applied Medical Research (CIMA), University of Navarra, Pamplona, Spain
| | | | - Ignacio Melero
- Division of Immunology and Immunotherapy, Center for Applied Medical Research (CIMA), University of Navarra, Pamplona, Spain.,Instituto de Investigación Sanitaria de Navarra (IdISNA), Pamplona, Spain.,University Clinic, University of Navarra, Pamplona, Spain
| |
Collapse
|
25
|
Bell RB, Leidner RS, Crittenden MR, Curti BD, Feng Z, Montler R, Gough MJ, Fox BA, Weinberg AD, Urba WJ. OX40 signaling in head and neck squamous cell carcinoma: Overcoming immunosuppression in the tumor microenvironment. Oral Oncol 2016; 52:1-10. [DOI: 10.1016/j.oraloncology.2015.11.009] [Citation(s) in RCA: 47] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2015] [Revised: 10/21/2015] [Accepted: 11/09/2015] [Indexed: 12/12/2022]
|
26
|
Tsurutani N, Mittal P, St Rose MC, Ngoi SM, Svedova J, Menoret A, Treadway FB, Laubenbacher R, Suárez-Ramírez JE, Cauley LS, Adler AJ, Vella AT. Costimulation Endows Immunotherapeutic CD8 T Cells with IL-36 Responsiveness during Aerobic Glycolysis. THE JOURNAL OF IMMUNOLOGY 2015; 196:124-34. [PMID: 26573834 DOI: 10.4049/jimmunol.1501217] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/28/2015] [Accepted: 10/17/2015] [Indexed: 01/07/2023]
Abstract
CD134- and CD137-primed CD8 T cells mount powerful effector responses upon recall, but even without recall these dual-costimulated T cells respond to signal 3 cytokines such as IL-12. We searched for alternative signal 3 receptor pathways and found the IL-1 family member IL-36R. Although IL-36 alone did not stimulate effector CD8 T cells, in combination with IL-12, or more surprisingly IL-2, it induced striking and rapid TCR-independent IFN-γ synthesis. To understand how signal 3 responses functioned in dual-costimulated T cells we showed that IL-2 induced IL-36R gene expression in a JAK/STAT-dependent manner. These data help delineate a sequential stimulation process where IL-2 conditioning must precede IL-36 for IFN-γ synthesis. Importantly, this responsive state was transient and functioned only in effector T cells capable of aerobic glycolysis. Specifically, as the effector T cells metabolized glucose and consumed O2, they also retained potential to respond through IL-36R. This suggests that T cells use innate receptor pathways such as the IL-36R/axis when programmed for aerobic glycolysis. To explore a function for IL-36R in vivo, we showed that dual costimulation therapy reduced B16 melanoma tumor growth while increasing IL-36R gene expression. In summary, cytokine therapy to eliminate tumors may target effector T cells, even outside of TCR specificity, as long as the effectors are in the correct metabolic state.
Collapse
Affiliation(s)
- Naomi Tsurutani
- Department of Immunology, University of Connecticut Health Center, Farmington, CT 06030; and
| | - Payal Mittal
- Department of Immunology, University of Connecticut Health Center, Farmington, CT 06030; and
| | - Marie-Clare St Rose
- Department of Immunology, University of Connecticut Health Center, Farmington, CT 06030; and
| | - Soo Mun Ngoi
- Department of Immunology, University of Connecticut Health Center, Farmington, CT 06030; and
| | - Julia Svedova
- Department of Immunology, University of Connecticut Health Center, Farmington, CT 06030; and
| | - Antoine Menoret
- Department of Immunology, University of Connecticut Health Center, Farmington, CT 06030; and
| | - Forrest B Treadway
- Center for Quantitative Medicine, School of Medicine, University of Connecticut Health Center, Farmington, CT 06030
| | - Reinhard Laubenbacher
- Center for Quantitative Medicine, School of Medicine, University of Connecticut Health Center, Farmington, CT 06030
| | - Jenny E Suárez-Ramírez
- Department of Immunology, University of Connecticut Health Center, Farmington, CT 06030; and
| | - Linda S Cauley
- Department of Immunology, University of Connecticut Health Center, Farmington, CT 06030; and
| | - Adam J Adler
- Department of Immunology, University of Connecticut Health Center, Farmington, CT 06030; and
| | - Anthony T Vella
- Department of Immunology, University of Connecticut Health Center, Farmington, CT 06030; and
| |
Collapse
|
27
|
Mittal P, St Rose MC, Wang X, Ryan JM, Wasser JS, Vella AT, Adler AJ. Tumor-Unrelated CD4 T Cell Help Augments CD134 plus CD137 Dual Costimulation Tumor Therapy. THE JOURNAL OF IMMUNOLOGY 2015; 195:5816-26. [PMID: 26561553 DOI: 10.4049/jimmunol.1502032] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/14/2015] [Accepted: 10/15/2015] [Indexed: 11/19/2022]
Abstract
The ability of immune-based cancer therapies to elicit beneficial CD8(+) CTLs is limited by tolerance pathways that inactivate tumor-specific CD4 Th cells. A strategy to bypass this problem is to engage tumor-unrelated CD4 Th cells. Thus, CD4 T cells, regardless of their specificity per se, can boost CD8(+) CTL priming as long as the cognate epitopes are linked via presentation on the same dendritic cell. In this study, we assessed the therapeutic impact of engaging tumor-unrelated CD4 T cells during dual costimulation with CD134 plus CD137 that provide help via the above-mentioned classical linked pathway, as well as provide nonlinked help that facilitates CTL function in T cells not directly responding to cognate Ag. We found that engagement of tumor-unrelated CD4 Th cells dramatically boosted the ability of dual costimulation to control the growth of established B16 melanomas. Surprisingly, this effect depended upon a CD134-dependent component that was extrinsic to the tumor-unrelated CD4 T cells, suggesting that the dual costimulated helper cells are themselves helped by a CD134(+) cell(s). Nevertheless, the delivery of therapeutic help tracked with an increased frequency of tumor-infiltrating granzyme B(+) effector CD8 T cells and a reciprocal decrease in Foxp3(+)CD4(+) cell frequency. Notably, the tumor-unrelated CD4 Th cells also infiltrated the tumors, and their deletion several days following initial T cell priming negated their therapeutic impact. Taken together, dual costimulation programs tumor-unrelated CD4 T cells to deliver therapeutic help during both the priming and effector stages of the antitumor response.
Collapse
Affiliation(s)
- Payal Mittal
- Department of Immunology, University of Connecticut School of Medicine, Farmington, CT 06030; and
| | - Marie-Clare St Rose
- Department of Immunology, University of Connecticut School of Medicine, Farmington, CT 06030; and
| | - Xi Wang
- Department of Immunology, University of Connecticut School of Medicine, Farmington, CT 06030; and
| | - Joseph M Ryan
- Department of Immunology, University of Connecticut School of Medicine, Farmington, CT 06030; and
| | - Jeffrey S Wasser
- The Carole and Ray Neag Comprehensive Cancer Center, University of Connecticut School of Medicine, Farmington, CT 06030
| | - Anthony T Vella
- Department of Immunology, University of Connecticut School of Medicine, Farmington, CT 06030; and
| | - Adam J Adler
- Department of Immunology, University of Connecticut School of Medicine, Farmington, CT 06030; and
| |
Collapse
|
28
|
Reuter D, Staege MS, Kühnöl CD, Föll J. Immunostimulation by OX40 Ligand Transgenic Ewing Sarcoma Cells. Front Oncol 2015; 5:242. [PMID: 26579494 PMCID: PMC4621427 DOI: 10.3389/fonc.2015.00242] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2015] [Accepted: 10/13/2015] [Indexed: 12/21/2022] Open
Abstract
Interleukin-2 (IL-2) transgenic Ewing sarcoma cells can induce tumor specific T and NK cell responses and reduce tumor growth in vivo and in vitro. Nevertheless, the efficiency of this stimulation is not high enough to inhibit tumor growth completely. In addition to recognition of the cognate antigen, optimal T-cell stimulation requires signals from so-called co-stimulatory molecules. Several members of the tumor necrosis factor superfamily have been identified as co-stimulatory molecules that can augment antitumor immune responses. OX40 (CD134) and OX40 ligand (OX40L = CD252; also known as tumor necrosis factor ligand family member 4) is one example of such receptor/ligand pair with co-stimulatory function. In the present investigation, we generated OX40L transgenic Ewing sarcoma cells and tested their immunostimulatory activity in vitro. OX40L transgenic Ewing sarcoma cells showed preserved expression of Ewing sarcoma-associated (anti)gens including lipase member I, cyclin D1 (CCND1), cytochrome P450 family member 26B1 (CYP26B1), and the Ewing sarcoma breakpoint region 1-friend leukemia virus integration 1 (EWSR1-FLI1) oncogene. OX40L-expressing tumor cells showed a trend for enhanced immune stimulation against Ewing sarcoma cells in combination with IL-2 and stimulation of CD137. Our data suggest that inclusion of the OX40/OX40L pathway of co-stimulation might improve immunotherapy strategies for the treatment of Ewing sarcoma.
Collapse
Affiliation(s)
- Dajana Reuter
- University Clinic and Polyclinic for Child and Adolescent Medicine, Martin Luther University Halle-Wittenberg , Halle , Germany
| | - Martin S Staege
- University Clinic and Polyclinic for Child and Adolescent Medicine, Martin Luther University Halle-Wittenberg , Halle , Germany
| | - Caspar D Kühnöl
- University Clinic and Polyclinic for Child and Adolescent Medicine, Martin Luther University Halle-Wittenberg , Halle , Germany
| | - Jürgen Föll
- University Clinic and Polyclinic for Child and Adolescent Medicine, Martin Luther University Halle-Wittenberg , Halle , Germany ; Department of Pediatric Hematology, Oncology and Stem Cell Transplantation, University Hospital Regensburg , Regensburg , Germany
| |
Collapse
|
29
|
Dushyanthen S, Beavis PA, Savas P, Teo ZL, Zhou C, Mansour M, Darcy PK, Loi S. Relevance of tumor-infiltrating lymphocytes in breast cancer. BMC Med 2015; 13:202. [PMID: 26300242 PMCID: PMC4547422 DOI: 10.1186/s12916-015-0431-3] [Citation(s) in RCA: 168] [Impact Index Per Article: 16.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/11/2015] [Accepted: 07/22/2015] [Indexed: 12/13/2022] Open
Abstract
While breast cancer has not been considered a cancer amenable to immunotherapeutic approaches, recent studies have demonstrated evidence of significant immune cell infiltration via tumor-infiltrating lymphocytes in a subset of patient tumors. In this review we present the current evidence highlighting the clinical relevance and utility of tumor-infiltrating lymphocytes in breast cancer. Retrospective and prospective studies have shown that the presence of tumor-infiltrating lymphocytes is a prognostic marker for higher responses to neoadjuvant chemotherapy and better survival, particularly in triple negative and HER2-positive early breast cancer. Further work is required to determine the immune subsets important in this response and to discover ways of encouraging immune infiltrate in tumor-infiltrating lymphocytes-negative patients.
Collapse
Affiliation(s)
- Sathana Dushyanthen
- Division of Research and Cancer Medicine, Peter MacCallum Cancer Centre, East Melbourne, VIC, Australia
| | - Paul A Beavis
- Division of Research and Cancer Medicine, Peter MacCallum Cancer Centre, East Melbourne, VIC, Australia
| | - Peter Savas
- Division of Research and Cancer Medicine, Peter MacCallum Cancer Centre, East Melbourne, VIC, Australia
| | - Zhi Ling Teo
- Division of Research and Cancer Medicine, Peter MacCallum Cancer Centre, East Melbourne, VIC, Australia
| | - Chenhao Zhou
- Division of Research and Cancer Medicine, Peter MacCallum Cancer Centre, East Melbourne, VIC, Australia
| | - Mariam Mansour
- Division of Research and Cancer Medicine, Peter MacCallum Cancer Centre, East Melbourne, VIC, Australia
| | - Phillip K Darcy
- Division of Research and Cancer Medicine, Peter MacCallum Cancer Centre, East Melbourne, VIC, Australia.,Sir Peter MacCallum Department of Oncology, The University of Melbourne, Parkville, VIC, Australia
| | - Sherene Loi
- Division of Research and Cancer Medicine, Peter MacCallum Cancer Centre, East Melbourne, VIC, Australia. .,Sir Peter MacCallum Department of Oncology, The University of Melbourne, Parkville, VIC, Australia. .,Peter MacCallum Cancer Centre, East Melbourne, VIC, Australia.
| |
Collapse
|
30
|
Abstract
The clinical benefit of therapeutic cancer vaccines has been established. Whereas regression of lesions was shown for premalignant lesions caused by HPV, clinical benefit in cancer patients was mostly noted as prolonged survival. Suboptimal vaccine design and an immunosuppressive cancer microenvironment are the root causes of the lack of cancer eradication. Effective cancer vaccines deliver concentrated antigen to both HLA class I and II molecules of DCs, promoting both CD4 and CD8 T cell responses. Optimal vaccine platforms include DNA and RNA vaccines and synthetic long peptides. Antigens of choice include mutant sequences, selected cancer testis antigens, and viral antigens. Drugs or physical treatments can mitigate the immunosuppressive cancer microenvironment and include chemotherapeutics, radiation, indoleamine 2,3-dioxygenase (IDO) inhibitors, inhibitors of T cell checkpoints, agonists of selected TNF receptor family members, and inhibitors of undesirable cytokines. The specificity of therapeutic vaccination combined with such immunomodulation offers an attractive avenue for the development of future cancer therapies.
Collapse
|
31
|
Yonezawa A, Dutt S, Chester C, Kim J, Kohrt HE. Boosting Cancer Immunotherapy with Anti-CD137 Antibody Therapy. Clin Cancer Res 2015; 21:3113-20. [PMID: 25908780 PMCID: PMC5422104 DOI: 10.1158/1078-0432.ccr-15-0263] [Citation(s) in RCA: 104] [Impact Index Per Article: 10.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2015] [Accepted: 03/17/2015] [Indexed: 11/16/2022]
Abstract
In the past 5 years, immunomodulatory antibodies have revolutionized cancer immunotherapy. CD137, a member of the tumor necrosis factor receptor superfamily, represents a promising target for enhancing antitumor immune responses. CD137 helps regulate the activation of many immune cells, including CD4(+) T cells, CD8(+) T cells, dendritic cells, and natural killer cells. Recent studies indicate that the antitumor efficacy of therapeutic tumor-targeting antibodies can be augmented by the addition of agonistic antibodies targeting CD137. As ligation of CD137 provides a costimulatory signal in multiple immune cell subsets, combination therapy of CD137 antibody with therapeutic antibodies and/or vaccination has the potential to improve cancer treatment. Recently, clinical trials of combination therapies with agonistic anti-CD137 mAbs have been launched. In this review, we discuss the recent advances and clinical promise of agonistic anti-CD137 monoclonal antibody therapy.
Collapse
Affiliation(s)
- Atsushi Yonezawa
- Division of Oncology, Department of Medicine, Stanford University, Stanford, California. Department of Clinical Pharmacology and Therapeutics, Kyoto University Hospital, Sakyo-ku, Kyoto, Japan
| | - Suparna Dutt
- Immunology and Rheumatology, Stanford University, Stanford, California
| | - Cariad Chester
- Division of Oncology, Department of Medicine, Stanford University, Stanford, California. Institute for Immunity, Transplantation and Infection, Stanford University, Stanford, California
| | - Jeewon Kim
- Transgenic, Knockout and Tumor Model Center, Stanford Cancer Institute, School of Medicine, Stanford University, Stanford, California
| | - Holbrook E Kohrt
- Division of Oncology, Department of Medicine, Stanford University, Stanford, California.
| |
Collapse
|
32
|
Lollini PL, Cavallo F, Nanni P, Quaglino E. The Promise of Preventive Cancer Vaccines. Vaccines (Basel) 2015; 3:467-89. [PMID: 26343198 PMCID: PMC4494347 DOI: 10.3390/vaccines3020467] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2015] [Revised: 06/04/2015] [Accepted: 06/08/2015] [Indexed: 01/01/2023] Open
Abstract
Years of unsuccessful attempts at fighting established tumors with vaccines have taught us all that they are only able to truly impact patient survival when used in a preventive setting, as would normally be the case for traditional vaccines against infectious diseases. While true primary cancer prevention is still but a long-term goal, secondary and tertiary prevention are already in the clinic and providing encouraging results. A combination of immunopreventive cancer strategies and recently approved checkpoint inhibitors is a further promise of forthcoming successful cancer disease control, but prevention will require a considerable reduction of currently reported toxicities. These considerations summed with the increased understanding of tumor antigens allow space for an optimistic view of the future.
Collapse
Affiliation(s)
- Pier-Luigi Lollini
- Department of Experimental Diagnostic and Specialty Medicine (DIMES), University of Bologna, Viale Filopanti 22, Bologna 40126, Italy.
| | - Federica Cavallo
- Department of Molecular Biotechnology and Health Sciences, Molecular Biotechnology Center, University of Torino, Via Nizza 52, Torino 10126, Italy.
| | - Patrizia Nanni
- Department of Experimental Diagnostic and Specialty Medicine (DIMES), University of Bologna, Viale Filopanti 22, Bologna 40126, Italy.
| | - Elena Quaglino
- Department of Molecular Biotechnology and Health Sciences, Molecular Biotechnology Center, University of Torino, Via Nizza 52, Torino 10126, Italy.
| |
Collapse
|
33
|
Bartkowiak T, Curran MA. 4-1BB Agonists: Multi-Potent Potentiators of Tumor Immunity. Front Oncol 2015; 5:117. [PMID: 26106583 PMCID: PMC4459101 DOI: 10.3389/fonc.2015.00117] [Citation(s) in RCA: 198] [Impact Index Per Article: 19.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2015] [Accepted: 05/11/2015] [Indexed: 01/12/2023] Open
Abstract
Immunotherapy is a rapidly expanding field of oncology aimed at targeting, not the tumor itself, but the immune system combating the cancerous lesion. Of the many approaches currently under study to boost anti-tumor immune responses; modulation of immune co-receptors on lymphocytes in the tumor microenvironment has thus far proven to be the most effective. Antibody blockade of the T cell co-inhibitory receptor cytotoxic T lymphocyte antigen-4 (CTLA-4) has become the first FDA approved immune checkpoint blockade; however, tumor infiltrating lymphocytes express a diverse array of additional stimulatory and inhibitory co-receptors, which can be targeted to boost tumor immunity. Among these, the co-stimulatory receptor 4-1BB (CD137/TNFSF9) possesses an unequaled capacity for both activation and pro-inflammatory polarization of anti-tumor lymphocytes. While functional studies of 4-1BB have focused on its prominent role in augmenting cytotoxic CD8 T cells, 4-1BB can also modulate the activity of CD4 T cells, B cells, natural killer cells, monocytes, macrophages, and dendritic cells. 4-1BB’s expression on both T cells and antigen presenting cells, coupled with its capacity to promote survival, expansion, and enhanced effector function of activated T cells, has made it an alluring target for tumor immunotherapy. In contrast to immune checkpoint blocking antibodies, 4-1BB agonists can both potentiate anti-tumor and anti-viral immunity, while at the same time ameliorating autoimmune disease. Despite this, 4-1BB agonists can trigger high grade liver inflammation which has slowed their clinical development. In this review, we discuss how the underlying immunobiology of 4-1BB activation suggests the potential for therapeutically synergistic combination strategies in which immune adverse events can be minimized.
Collapse
Affiliation(s)
- Todd Bartkowiak
- Department of Immunology, University of Texas MD Anderson Cancer Center , Houston, TX , USA ; The University of Texas Graduate School of Biomedical Sciences at Houston , Houston, TX , USA
| | - Michael A Curran
- Department of Immunology, University of Texas MD Anderson Cancer Center , Houston, TX , USA ; The University of Texas Graduate School of Biomedical Sciences at Houston , Houston, TX , USA
| |
Collapse
|
34
|
Linch SN, McNamara MJ, Redmond WL. OX40 Agonists and Combination Immunotherapy: Putting the Pedal to the Metal. Front Oncol 2015; 5:34. [PMID: 25763356 PMCID: PMC4329814 DOI: 10.3389/fonc.2015.00034] [Citation(s) in RCA: 183] [Impact Index Per Article: 18.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2014] [Accepted: 01/30/2015] [Indexed: 12/24/2022] Open
Abstract
Recent studies have highlighted the therapeutic efficacy of immunotherapy, a class of cancer treatments that utilize the patient’s own immune system to destroy cancerous cells. Within a tumor the presence of a family of negative regulatory molecules, collectively known as “checkpoint inhibitors,” can inhibit T cell function to suppress anti-tumor immunity. Checkpoint inhibitors, such as CTLA-4 and PD-1, attenuate T cell proliferation and cytokine production. Targeted blockade of CTLA-4 or PD-1 with antagonist monoclonal antibodies (mAbs) releases the “brakes” on T cells to boost anti-tumor immunity. Generating optimal “killer” CD8 T cell responses also requires T cell receptor activation plus co-stimulation, which can be provided through ligation of tumor necrosis factor receptor family members, including OX40 (CD134) and 4-1BB (CD137). OX40 is of particular interest as treatment with an activating (agonist) anti-OX40 mAb augments T cell differentiation and cytolytic function leading to enhanced anti-tumor immunity against a variety of tumors. When used as single agents, these drugs can induce potent clinical and immunologic responses in patients with metastatic disease. However, each of these agents only benefits a subset of patients, highlighting the critical need for more effective combinatorial therapeutic strategies. In this review, we will discuss our current understanding of the cellular and molecular mechanisms by which OX40 agonists synergize with checkpoint inhibitor blockade to augment T cell-mediated anti-tumor immunity and the potential opportunities for clinical translation of combinatorial immunotherapeutic strategies.
Collapse
Affiliation(s)
- Stefanie N Linch
- Robert W. Franz Cancer Research Center, Earle A. Chiles Research Institute, Providence Portland Medical Center , Portland, OR , USA
| | - Michael J McNamara
- Robert W. Franz Cancer Research Center, Earle A. Chiles Research Institute, Providence Portland Medical Center , Portland, OR , USA
| | - William L Redmond
- Robert W. Franz Cancer Research Center, Earle A. Chiles Research Institute, Providence Portland Medical Center , Portland, OR , USA
| |
Collapse
|
35
|
Adler AJ, Vella AT. Betting on improved cancer immunotherapy by doubling down on CD134 and CD137 co-stimulation. Oncoimmunology 2014; 2:e22837. [PMID: 23482891 PMCID: PMC3583935 DOI: 10.4161/onci.22837] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
The ability of T cells to recognize a vast array of antigens enables them to destroy tumor cells while inflicting minimal collateral damage. Nevertheless, tumor antigens often are a form of self-antigen, and thus tumor immunity can be dampened by tolerance mechanisms that evolved to prevent autoimmunity. Since tolerance can be induced by steady-state antigen-presenting cells that provide insufficient co-stimulation, the exogenous administration of co-stimulatory agonists can favor the expansion and tumoricidal functions of tumor-specific T cells. Agonists of the co-stimulatory tumor necrosis factor receptor (TNFR) family members CD134 and CD137 exert antitumor activity in mice, and as monotherapies have exhibited encouraging results in clinical trials. This review focuses on how the dual administration of CD134 and CD137 agonists synergistically boosts T-cell priming and elaborates a multi-pronged antitumor immune response, as well as how such dual co-stimulation might be translated into effective anticancer therapies.
Collapse
Affiliation(s)
- Adam J Adler
- Department of Immunology; University of Connecticut Health Center; Farmington, CT USA
| | | |
Collapse
|
36
|
Sallin MA, Zhang X, So EC, Burch E, Cai L, Lin W, Chapoval AI, Strome SE. The anti-lymphoma activities of anti-CD137 monoclonal antibodies are enhanced in FcγRIII(-/-) mice. Cancer Immunol Immunother 2014; 63:947-58. [PMID: 24927849 PMCID: PMC11029484 DOI: 10.1007/s00262-014-1567-2] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2013] [Accepted: 05/30/2014] [Indexed: 11/25/2022]
Abstract
Agonistic monoclonal antibodies (mAbs) directed against the co-signaling molecule CD137 (4-1BB) elicit potent anti-tumor immunity in mice. This anti-tumor immunity has traditionally been thought to result from the ability of the Fab portion of anti-CD137 to function as an analog for CD137L. Although binding of CD137 by anti-CD137 mAbs has the potential to cross-link the Fc fragments, enabling Fc engagement of low to moderate affinity Fc gamma receptors (FcγR), the relative import of such Fc-FcγR interactions in mediating anti-CD137 associated anti-tumor immunity is unknown. We studied the ability of a rat anti-mouse CD137 mAb (2A) to mediate the anti-tumor response against the EL4E7 lymphoma in WT and FcγR(-/-) strains. 2A-treated FcRγ(-/-) mice had improved anti-tumor immunity against EL4E7, which could be completely recapitulated in FcγRIII(-/-) animals. These improved anti-tumor responses were associated with increased splenic CD8β T cell and dendritic cell (DC) populations. Furthermore, there was an increase in the number of DCs expressing high levels of the CD40, CD80, and CD86 molecules that are associated with more effective antigen presentation. Our results demonstrate an unexpected inhibitory role for FcγRIII in the anti-tumor function of anti-CD137 and underscore the need to consider antibody isotype when engineering therapeutic mAbs.
Collapse
MESH Headings
- Animals
- Antibodies, Immobilized/immunology
- Antibodies, Immobilized/metabolism
- Antibodies, Monoclonal/immunology
- Antibodies, Monoclonal/metabolism
- Antibodies, Monoclonal/pharmacology
- Female
- HEK293 Cells
- Humans
- Lymphoma/immunology
- Lymphoma/therapy
- Melanoma, Experimental/immunology
- Melanoma, Experimental/therapy
- Mice
- Mice, Inbred C57BL
- Random Allocation
- Rats
- Receptors, IgG/deficiency
- Receptors, IgG/immunology
- Receptors, IgG/metabolism
- Transfection
- Tumor Necrosis Factor Receptor Superfamily, Member 9/biosynthesis
- Tumor Necrosis Factor Receptor Superfamily, Member 9/genetics
- Tumor Necrosis Factor Receptor Superfamily, Member 9/immunology
Collapse
Affiliation(s)
- Michelle A. Sallin
- Department of Otorhinolaryngology-Head and Neck Surgery, University of Maryland School of Medicine, 16 South Eutaw St. Suite 500, Baltimore, MD 21201-168 USA
| | - Xiaoyu Zhang
- Department of Otorhinolaryngology-Head and Neck Surgery, University of Maryland School of Medicine, 16 South Eutaw St. Suite 500, Baltimore, MD 21201-168 USA
| | - Edward C. So
- Department of Microbiology and Immunology, University of Maryland School of Medicine, 855 West Baltimore Street, Suite 380, HSF-I, Baltimore, MD 21201 USA
| | - Erin Burch
- Department of Otorhinolaryngology-Head and Neck Surgery, University of Maryland School of Medicine, 16 South Eutaw St. Suite 500, Baltimore, MD 21201-168 USA
| | - Ling Cai
- Department of Biostatistics, Bioinformatics and Biomathematics, Georgetown University Medical Center, 4000 Reservoir Road, NW, Building D-103, Washington, DC 20057 USA
| | - Wei Lin
- College of Rehabilitation Medicine, Fujian University of Traditional Chinese Medicine, Fuzhou, 350003 Fujian China
| | - Andrei I. Chapoval
- Russian-American Anti-Cancer Center, Department of Physico-Chemical Biology and Biotechnology, Altai State University, 61 Lenin Street, 656049 Barnaul, Altai Territory Russia
| | - Scott E. Strome
- Department of Otorhinolaryngology-Head and Neck Surgery, University of Maryland School of Medicine, 16 South Eutaw St. Suite 500, Baltimore, MD 21201-168 USA
| |
Collapse
|
37
|
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: 1.5] [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
|
38
|
Zhong R, Han B, Zhong H. A prospective study of the efficacy of a combination of autologous dendritic cells, cytokine-induced killer cells, and chemotherapy in advanced non-small cell lung cancer patients. Tumour Biol 2013; 35:987-94. [PMID: 24006222 DOI: 10.1007/s13277-013-1132-1] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2013] [Accepted: 08/23/2013] [Indexed: 12/13/2022] Open
Abstract
Dendritic cells (DC) play a crucial role in the induction of an effective antitumor immune response. Cytokine-induced killer (CIK) cells, a subset of T lymphocytes, have the capacity to eliminate cancer cells. This study was to evaluate the correlation between the frequency of DC/CIK immunotherapies following regular chemotherapy, the time-to-progression (TTP), and overall survival (OS) of advanced non-small lung cancer patients. Sixty patients with IIIB-IV non-small-cell lung carcinoma (NSCLC) were enrolled from August 2007 to December 2009 and were randomized into two groups. All 60 patients received four courses of navelbine-platinum (NP) chemotherapy. In one group, 30 patients were treated with adoptive autologous DC/CIK cell transfusion twice every 30 days. In the other group, the patients received immunotherapies more than twice every 30 days. The adverse effects, TTP, and OS were evaluated between the two groups. Median survival time of all 60 patients was 13.80 months. The 1-, 2-, and 3-year overall survival rates were 60.0, 21.7, and 15.0 %, respectively. The 1-, 2-, and 3-year overall survival rates of patients receiving more than two immunotherapies were 63.3, 30.0, and 23.3 %, and the rates of those receiving two immunotherapies were 56.7, 13.3, and 6.7 %, respectively. The difference between the two groups was statistically significant (P = 0.037). Compared with patients in the fewer immunotherapies group, TTP in the group receiving more immunotherapies significantly prolonged, with the median improving from 6.2 months (95 % CI, 5.35-9.24) to 7.3 months (95 % CI, 5.45-6.95; P = 0.034). The adverse effects of chemoimmunotherapy were tolerable. Advanced NSCLC patients can benefit from the combination of DC/CIK immunotherapies following conventional chemotherapy. More than two immunotherapies improved TTP and OS of those patients in this study.
Collapse
Affiliation(s)
- Runbo Zhong
- Department of Pulmonary Disease, Shanghai Chest Hospital, Shanghai Jiao Tong University, No. 241, Huaihai Road (W), Shanghai, People's Republic of China
| | | | | |
Collapse
|
39
|
CD134/CD137 dual costimulation-elicited IFN-γ maximizes effector T-cell function but limits Treg expansion. Immunol Cell Biol 2013; 91:173-83. [PMID: 23295363 PMCID: PMC3570742 DOI: 10.1038/icb.2012.74] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
T cell tolerance to tumor antigens represents a major hurdle in generating tumor immunity. Combined administration of agonistic monoclonal antibodies to the costimulatory receptors CD134 plus CD137 can program T cells responding to tolerogenic antigen to undergo expansion and effector T cell differentiation, and also elicits tumor immunity. Nevertheless, CD134 and CD137 agonists can also engage inhibitory immune components. To understand how immune stimulatory versus inhibitory components are regulated during CD134 plus CD137 dual costimulation, the current study utilized a model where dual costimulation programs T cells encountering a highly tolerogenic self-antigen to undergo effector differentiation. IFN-γ was found to play a pivotal role in maximizing the function of effector T cells while simultaneously limiting the expansion of CD4+CD25+Foxp3+ Tregs. In antigen-responding effector T cells, IFN-γ operates via a direct cell-intrinsic mechanism to cooperate with IL-2 to program maximal expression of granzyme B. Simultaneously, IFN-γ limits expression of the IL-2 receptor alpha chain (CD25) and IL-2 signaling through a mechanism that does not involve T-bet-mediated repression of IL-2. IFN-γ also limited CD25 and Foxp3 expression on bystanding CD4+Foxp3+ Tregs, and limited the potential of these Tregs to expand. These effects could not be explained by the ability of IFN-γ to limit IL-2 availability. Taken together, during dual costimulation IFN-γ interacts with IL-2 through distinct mechanisms to program maximal expression of effector molecules in antigen-responding T cells while simultaneously limiting Treg expansion.
Collapse
|
40
|
Abstract
Cancer vaccines have shown success in curing tumors in preclinical models. Accumulating evidence also supports their ability to induce immune responses in patients. In many cases, these responses correlate with improved clinical outcomes. However, cancer vaccines have not yet demonstrated their true potential in clinical trials. This is likely due to the difficulty in mounting a significant anti-tumor response in patients with advanced disease because of pre-existing tolerance mechanisms that are actively turning off immune recognition in cancer patients. This review will examine the recent progress being made in the design and implementation of whole cell cancer vaccines, one vaccine approach that simultaneously targets multiple tumor antigens to activate the immune response. These vaccines have been shown to induce antigen-specific T-cell responses. Preclinical studies evaluating these vaccines given in sequence with other agents and cancer treatment modalities support the use of immunomodulating doses of chemotherapy and radiation, as well as immune-modulating pathway-targeted monoclonal antibodies, to enhance the efficacy of cancer vaccines. Based on emerging preclinical data, clinical trials are currently exploring the use of combinatorial immune-based therapies for the treatment of cancer.
Collapse
Affiliation(s)
- Bridget P Keenan
- Graduate Program in Immunology, Johns Hopkins School of Medicine, Baltimore, MD, USA
| | | |
Collapse
|
41
|
Kroesen M, Lindau D, Hoogerbrugge P, Adema GJ. Immunocombination therapy for high-risk neuroblastoma. Immunotherapy 2012; 4:163-74. [PMID: 22394368 DOI: 10.2217/imt.11.169] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023] Open
Abstract
Neuroblastoma (NBL) is an aggressive malignancy of the sympathetic nervous system. Advanced-stage NBLs prove fatal in approximately 50% of patients within 5 years. Therefore, new treatment modalities are urgently needed. Immunotherapy is a treatment modality that can be combined with established forms of treatment. Administration of monoclonal antibodies or dendritic cell-based therapies alone can lead to favorable clinical outcomes in individual cancer patients; for example patients with melanoma, lymphoma and NBL. However, clinical benefit is still limited to a minority of patients, and further improvements are clearly needed. In this article, we review the most commonly used approaches to treat patients with NBL and highlight the prerequisites and opportunities of cell-based immunotherapy, involving both innate and adaptive immune-effector cells. Furthermore, we discuss the potential of the combined application of immunotherapy and novel tumor-targeted therapies for the treatment of both cancer in general and NBL in particular.
Collapse
Affiliation(s)
- Michiel Kroesen
- Department of Tumor Immunology, Nijmegen Centre for Molecular Life Sciences/278 TIL, 6500 HB Nijmegen, The Netherlands
| | | | | | | |
Collapse
|
42
|
Targeting costimulatory molecules to improve antitumor immunity. J Biomed Biotechnol 2012; 2012:926321. [PMID: 22500111 PMCID: PMC3303883 DOI: 10.1155/2012/926321] [Citation(s) in RCA: 62] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2011] [Revised: 10/12/2011] [Accepted: 11/16/2011] [Indexed: 12/12/2022] Open
Abstract
The full activation of T cells necessitates the concomitant activation of two signals, the engagement of T-cell receptor by peptide/major histocompatibility complex II and an additional signal delivered by costimulatory molecules. The best characterized costimulatory molecules belong to B7/CD28 and TNF/TNFR families and play crucial roles in the modulation of immune response and improvement of antitumor immunity. Unfortunately, tumors often generate an immunosuppressive microenvironment, where T-cell response is attenuated by the lack of costimulatory molecules on the surface of cancer cells. Thus, targeting costimulatory pathways represent an attractive therapeutic strategy to enhance the antitumor immunity in several human cancers. Here, latest therapeutic approaches targeting costimulatory molecules will be described.
Collapse
|
43
|
Qui HZ, Hagymasi AT, Bandyopadhyay S, St Rose MC, Ramanarasimhaiah R, Ménoret A, Mittler RS, Gordon SM, Reiner SL, Vella AT, Adler AJ. CD134 plus CD137 dual costimulation induces Eomesodermin in CD4 T cells to program cytotoxic Th1 differentiation. THE JOURNAL OF IMMUNOLOGY 2011; 187:3555-64. [PMID: 21880986 DOI: 10.4049/jimmunol.1101244] [Citation(s) in RCA: 125] [Impact Index Per Article: 8.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Cytotoxic CD4 Th1 cells are emerging as a therapeutically useful T cell lineage that can effectively target tumors, but until now the pathways that govern their differentiation have been poorly understood. We demonstrate that CD134 (OX40) costimulation programs naive self- and virus-reactive CD4 T cells to undergo in vivo differentiation into cytotoxic Th1 effectors. CD137 (4-1BB) costimulation maximized clonal expansion, and IL-2 was necessary for cytotoxic Th1 differentiation. Importantly, the T-box transcription factor Eomesodermin was critical for inducing the cytotoxic marker granzyme B. CD134 plus CD137 dual costimulation also imprinted a cytotoxic phenotype on bystanding CD4 T cells. Thus, to our knowledge, the current study identifies for the first time a specific costimulatory pathway and an intracellular mechanism relying on Eomesodermin that induces both Ag-specific and bystander cytotoxic CD4 Th1 cells. This mechanism might be therapeutically useful because CD134 plus CD137 dual costimulation induced CD4 T cell-dependent tumoricidal function in a mouse melanoma model.
Collapse
Affiliation(s)
- Harry Z Qui
- Department of Immunology, University of Connecticut Health Center, Farmington, CT 06030, USA
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
44
|
Smith SE, Hoelzinger DB, Dominguez AL, Van Snick J, Lustgarten J. Signals through 4-1BB inhibit T regulatory cells by blocking IL-9 production enhancing antitumor responses. Cancer Immunol Immunother 2011; 60:1775-87. [PMID: 21789593 DOI: 10.1007/s00262-011-1075-6] [Citation(s) in RCA: 50] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2011] [Accepted: 07/11/2011] [Indexed: 12/25/2022]
Abstract
Previous studies from our laboratory indicate that intratumoral (i.t.) injections of CpG-ODN are the most effective adjuvant strategy to induce an antitumor immune response in tolerant BALB-neuT mice but insufficient for tumor eradication. We evaluated whether this treatment strategy could be enhanced by the presence of anti-OX40 and anti-4-1BB antibodies. Treatment with anti-4-1BB resulted in a greater antitumor response than anti-OX40. The results indicate that anti-4-1BB but not anti-OX40 inhibited the suppressive function of T regulatory cells (Tregs). Through microarray analysis we evaluated the mechanism by which anti-4-1BB inhibits iTregs using the Foxp3-GFP mice. We observed specific transcriptional differences in over 100 genes in iTregs treated with anti-4-1BB, and selected those genes that remained unaffected by exposure to anti-OX40. Interleukin 9 was transcriptionally down-regulated 28-fold by anti-4-1BB treatment, and this was matched by a significant reduction of IL-9 secretion by iTregs. Furthermore, blockade of the common γ-chain receptor resulted in the inhibition of iTreg-suppressive function. More importantly, neutralization of IL-9 plus i.t. injections of CpG-ODN induces tumor rejection in BALB-neuT and MUC-1 tolerant transgenic mice. These results indicate that IL-9 plays a role in iTreg biology during the tumor inflammatory process enhancing/promoting the suppressive function of these cells and that the blockade of IL-9 could serve as a novel strategy to modulate the function of Tregs to enhance the antitumor effect of tumor vaccines.
Collapse
Affiliation(s)
- Shannon E Smith
- Department of Immunology, Cancer Center Scottsdale, Mayo Clinic Arizona, Mayo Clinic College of Medicine, 13400 East Shea Boulevard, Scottsdale, AZ 85259, USA
| | | | | | | | | |
Collapse
|
45
|
Pardee AD, Wesa AK, Storkus WJ. Integrating costimulatory agonists to optimize immune-based cancer therapies. Immunotherapy 2010; 1:249-64. [PMID: 20046961 DOI: 10.2217/1750743x.1.2.249] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
While immunotherapy for cancer has become increasingly popular, clinical benefits for such approaches remain limited. This is likely due to tumor-associated immune suppression, particularly in the advanced-disease setting. Thus, a major goal of novel immunotherapeutic design has become the coordinate reversal of existing immune dysfunction and promotion of specific tumoricidal T-cell function. Costimulatory members of the TNF-receptor family are important regulators of T-cell-mediated immunity. Notably, agonist ligation of these receptors restores potent antitumor immunity in the tumor-bearing host. Current Phase I/II evaluation of TNF-receptor agonists as single-modality therapies will illuminate their safety, mechanism(s) of action, and best use in prospective combinational immunotherapy approaches capable of yielding superior benefits to cancer patients.
Collapse
Affiliation(s)
- Angela D Pardee
- University of Pittsburgh School of Medicine, PA, Pittsburgh, USA
| | | | | |
Collapse
|
46
|
Dubrot J, Portero A, Orive G, Hernández RM, Palazón A, Rouzaut A, Perez-Gracia JL, Hervás-Stubbs S, Pedraz JL, Melero I. Delivery of immunostimulatory monoclonal antibodies by encapsulated hybridoma cells. Cancer Immunol Immunother 2010; 59:1621-31. [PMID: 20607237 PMCID: PMC11030103 DOI: 10.1007/s00262-010-0888-z] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2009] [Accepted: 06/12/2010] [Indexed: 12/15/2022]
Abstract
Immunostimulatory monoclonal antibodies are immunoglobulins directed toward surface proteins of immune system cells that augment the immune response against cancer in a novel therapeutic fashion. Exogenous administration of the recombinant humanized immunoglobulins is being tested in clinical trials with agents of this kind directed at a variety of immune-controlling molecular targets. In this study, the encapsulation of antibody-producing hybridoma cells was tested in comparison with the systemic administration of monoclonal antibodies. Hybridomas producing anti-CD137 and anti-OX40 mAb were encapsulated in alginate to generate microcapsules containing viable cells that secrete antibody. Immobilized cells in vitro were able to release the rat immunoglobulin produced by the hybridomas into the supernatant. Microcapsules were implanted by injection into the subcutaneous tissue of mice and thereby provided a platform for viable secreting cells, which lasted for more than 1 week. The pharmacokinetic profile of the rat monoclonal antibodies following microcapsule implantation was similar to that attained following an intraperitoneal administration of the purified antibodies. The rat-mouse hybridoma cells did not engraft as tumors in immunocompetent mice, while they lethally xenografted in immunodeficient mice, if not microencapsulated. The antitumor therapeutic activity of the strategy was studied on established CT26 colon carcinomas resulting in complete tumor eradication in an elevated fraction of cases and strong tumor-specific CTL responses with either anti-CD137 or anti-OX40 producing hybridomas, thus offering proof of the concept. This form of administration permitted combinations of more than one immunostimulatory monoclonal antibody to exploit the synergistic effects such as those known to be displayed by anti-CD137 and anti-OX40 mAb.
Collapse
Affiliation(s)
- Juan Dubrot
- Centro de Investigación Médica Aplicada (CIMA), University of Navarra, CIMA. Av. Pio XII, 55 31008 Pamplona, Spain
| | - Aitziber Portero
- Laboratory of Pharmacy and Pharmaceutical Technology, Networking Biomedical Research Center on Bioengineering, Biomaterials and Nanomedicine, CIBER-BBN, SLFPB-EHU, Faculty of Pharmacy, University of the Basque Country, 01006 Vitoria-Gasteiz, Spain
| | - Gorka Orive
- Laboratory of Pharmacy and Pharmaceutical Technology, Networking Biomedical Research Center on Bioengineering, Biomaterials and Nanomedicine, CIBER-BBN, SLFPB-EHU, Faculty of Pharmacy, University of the Basque Country, 01006 Vitoria-Gasteiz, Spain
| | - Rosa María Hernández
- Laboratory of Pharmacy and Pharmaceutical Technology, Networking Biomedical Research Center on Bioengineering, Biomaterials and Nanomedicine, CIBER-BBN, SLFPB-EHU, Faculty of Pharmacy, University of the Basque Country, 01006 Vitoria-Gasteiz, Spain
| | - Asis Palazón
- Centro de Investigación Médica Aplicada (CIMA), University of Navarra, CIMA. Av. Pio XII, 55 31008 Pamplona, Spain
| | - Ana Rouzaut
- Centro de Investigación Médica Aplicada (CIMA), University of Navarra, CIMA. Av. Pio XII, 55 31008 Pamplona, Spain
| | | | - Sandra Hervás-Stubbs
- Centro de Investigación Médica Aplicada (CIMA), University of Navarra, CIMA. Av. Pio XII, 55 31008 Pamplona, Spain
| | - Jose Luis Pedraz
- Laboratory of Pharmacy and Pharmaceutical Technology, Networking Biomedical Research Center on Bioengineering, Biomaterials and Nanomedicine, CIBER-BBN, SLFPB-EHU, Faculty of Pharmacy, University of the Basque Country, 01006 Vitoria-Gasteiz, Spain
| | - Ignacio Melero
- Centro de Investigación Médica Aplicada (CIMA), University of Navarra, CIMA. Av. Pio XII, 55 31008 Pamplona, Spain
- Clinica Universitaria, Universidad de Navarra, Pamplona, Spain
| |
Collapse
|
47
|
Wang X, Zhou K, Huang L, Yan Y. Induction of anti-tumor immunity by dendritic cells pulsed with an endoplasmic reticulum retrieval signal modifies heparanase epitope in mice. Cytotherapy 2010; 12:735-42. [DOI: 10.3109/14653241003615156] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
|
48
|
Kuang Y, Weng X, Liu X, Zhu H, Chen Z, Jiang B, Chen H. Anti-tumor immune response induced by dendritic cells transduced with truncated PSMA IRES 4-1BBL recombinant adenoviruses. Cancer Lett 2010; 293:254-62. [PMID: 20149524 DOI: 10.1016/j.canlet.2010.01.011] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2009] [Revised: 12/28/2009] [Accepted: 01/17/2010] [Indexed: 10/19/2022]
Abstract
Up-regulation of receptor-ligand pairs during interaction of a peptide-bound MHC complex on dendritic cells (DCs) with cognate TCR may amplify, sustain, and drive diversity in the ensuing T cell immune response. Members of the TNF ligand superfamily and the TNFR superfamily contribute to this costimulatory molecule signaling. In the present study, we used replication deficient adenoviruses to introduce a tumor-associated Ag (a truncated human prostate-specific membrane antigen (tPSMA)) and the T cell costimulatory molecule 4-1BBL into murine DCs, and observed the ability of these recombinant DCs to elicit tPSMA-directed T-cell responses in vitro and anti-tumor immunity to RM-1-tPSMA in a murine tumor model. Infection of DCs with Ad-tPSMA-IRES-m4-1BBL induced tPSMA-specific proliferative responses and up-regulated CD80 and CD86 s signaling molecules. The cytotoxic T lymphocytes activated by the Ad-tPSMA-IRES-m4-1BBL-transfected DCs showed significantly higher IFN-gamma production and cytotoxicity against the RM-1 cells transfected with tPSMA. Moreover, vaccination of mice with Ad-tPSMA-IRES-m4-1BBL-transfected DCs induced a potent protective and therapeutic anti-tumor immunity to RM-1-tPSMA in a tumor model. These results demonstrated that development of DCs engineered to express tPSMA and 4-1BBL by recombinant adenovirus-mediated gene transfer may offer a new strategy for prostate cancer immunotherapy.
Collapse
Affiliation(s)
- Youlin Kuang
- Department of Urology, Renmin Hospital of Wuhan University, Wuhan University, Wuhan, China
| | | | | | | | | | | | | |
Collapse
|
49
|
|
50
|
Lustgarten J. Cancer, aging and immunotherapy: lessons learned from animal models. Cancer Immunol Immunother 2009; 58:1979-89. [PMID: 19238382 PMCID: PMC11030962 DOI: 10.1007/s00262-009-0677-8] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2008] [Accepted: 01/30/2009] [Indexed: 01/06/2023]
Abstract
Aging of the immune system is associated with a dramatic reduction in responsiveness as well as functional dysregulation. This deterioration of immune function with advancing age is associated with an increased incidence of cancer. Although there is a plethora of reports evaluating the effect of immunotherapy in stimulating antitumor immune responses, the majority of these studies do not pay attention to the effect aging has on the immune system. Studies from our group and others indicate that immunotherapies could be effective in the young, are not necessarily effective in the old. To optimally stimulate an antitumor immune response in the old, it is necessary to (1) identify and understand the intrinsic defects of the old immune system and (2) use relevant models that closely reflect those of cancer patients, where self-tolerance and aging are present simultaneously. The present review summarizes some defects found in the old immune system affecting the activation of antitumor immune responses, the strategies used to activate stronger antitumor immune response in the old and the use of a tolerant animal tumor model to target a self-tumor antigen for the optimization of immunotherapeutic interventions in the old.
Collapse
Affiliation(s)
- Joseph Lustgarten
- Department of Immunology, Mayo Clinic Arizona, Mayo Clinic College of Medicine, Scottsdale, AZ 85259, USA.
| |
Collapse
|