51
|
Shablak A, Hawkins RE, Rothwell DG, Elkord E. T cell-based immunotherapy of metastatic renal cell carcinoma: modest success and future perspective. Clin Cancer Res 2009; 15:6503-10. [PMID: 19843660 DOI: 10.1158/1078-0432.ccr-09-1605] [Citation(s) in RCA: 31] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Metastatic renal cell carcinoma (MRCC) remains a challenging malignancy to treat. Cancer immunotherapies have been extensively explored in melanoma and RCC as they poorly respond to conventional cytotoxic agents but show responses to a variety of immunologic agents. The recent considerable success of T cell-based immunotherapy in melanoma warrants further efforts to apply this treatment to other cancers including MRCC. Although RCC is an immunosensitive cancer, similar attempts in MRCC have shown a very limited success. In this review, we summarize the clinical data on T cell-based immunotherapies for MRCC showing the modest success that has been achieved to date. More importantly, we discuss potential strategies for improving its efficacy for the treatment of MRCC in light of the important achievements for treating metastatic melanoma. In particular, the growing evidence of success by combining expanded tumor-infiltrating lymphocytes with lymphodepletion merits investigation in MRCC. Identifying new RCC-associated antigens, optimized methods, and conditions for detection, isolation, and/or modification and expansion of tumor-specific T cells are all important strategies to be pursued for improving T cell-based immunotherapy of MRCC.
Collapse
Affiliation(s)
- Alaaeldin Shablak
- Department of Medical Oncology, School of Cancer, Enabling Sciences and Technology, The University of Manchester, Manchester Academic Health Science Centre, The Christie NHS Foundation Trust, Manchester, United Kingdom
| | | | | | | |
Collapse
|
52
|
Abstract
Adoptive T cell therapies can produce objective clinical responses in patients with hematologic and solid malignancies. Genetic manipulation of T lymphocytes has been proposed as a means of increasing the potency and range of this anti-tumor activity. We now review how coupling expression of transgenic receptors with countermeasures against potent tumor immune evasion strategies is proving highly effective in pre-clinical models and describe how these approaches are being evaluated in human subjects.
Collapse
Affiliation(s)
- Juan F. Vera
- Center for Cell and Gene Therapy, Baylor College of Medicine, The Methodist Hospital and Texas Children’s Hospital, Houston, TX, 77030, USA
- Department of Medicine, Baylor College of Medicine, The Methodist Hospital and Texas Children’s Hospital, Houston, TX, 77030, USA
| | - Malcolm K. Brenner
- Center for Cell and Gene Therapy, Baylor College of Medicine, The Methodist Hospital and Texas Children’s Hospital, Houston, TX, 77030, USA
- Department of Medicine, Baylor College of Medicine, The Methodist Hospital and Texas Children’s Hospital, Houston, TX, 77030, USA
- Department of Pediatrics, Baylor College of Medicine, The Methodist Hospital and Texas Children’s Hospital, Houston, TX, 77030, USA
| | - Gianpietro Dotti
- Center for Cell and Gene Therapy, Baylor College of Medicine, The Methodist Hospital and Texas Children’s Hospital, Houston, TX, 77030, USA
- Department of Medicine, Baylor College of Medicine, The Methodist Hospital and Texas Children’s Hospital, Houston, TX, 77030, USA
- Department of Immunology, Baylor College of Medicine, The Methodist Hospital and Texas Children’s Hospital, Houston, TX, 77030, USA
| |
Collapse
|
53
|
Geiger C, Nössner E, Frankenberger B, Falk CS, Pohla H, Schendel DJ. Harnessing innate and adaptive immunity for adoptive cell therapy of renal cell carcinoma. J Mol Med (Berl) 2009; 87:595-612. [PMID: 19271159 DOI: 10.1007/s00109-009-0455-2] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2008] [Revised: 02/18/2009] [Accepted: 02/20/2009] [Indexed: 12/22/2022]
Abstract
The development of immunotherapies for renal cell carcinoma (RCC) has been the subject of research for several decades. In addition to cytokine therapy, the benefit of various adoptive cell therapies has again come into focus in the past several years. Nevertheless, success in fighting this immunogenic tumor is still disappointing. RCC can attract a multitude of different effector cells of both the innate and adaptive immune system, including natural killer (NK) cells, gammadelta T cells, NK-like T cells, peptide-specific T cells, dendritic cells (DC), and regulatory T cells (Tregs). Based on intensive research on the biology and function of different immune cells, we now understand that individual cell types do not act in isolation but function within a complex network of intercellular interactions. These interactions play a pivotal role in the efficient activation and function of effector cells, which is a prerequisite for successful tumor elimination. This review provides a current overview of the diversity of effector cells having the capacity to recognize RCC. Aspects of the functions and anti-tumor properties that make them attractive candidates for adoptive cell therapies, as well as experience in clinical application are discussed. Improved knowledge of the biology of this immune network may help us to effectively harness various effector cells, placing us in a better position to develop new therapeutic strategies to successfully fight RCC.
Collapse
Affiliation(s)
- Christiane Geiger
- Institut für Molekulare Immunologie, Helmholtz Zentrum München, Deutsches Forschungszentrum für Gesundheit und Umwelt, Germany.
| | | | | | | | | | | |
Collapse
|
54
|
Liu Y, Peng Y, Mi M, Guevara-Patino J, Munn DH, Fu N, He Y. Lentivector immunization stimulates potent CD8 T cell responses against melanoma self-antigen tyrosinase-related protein 1 and generates antitumor immunity in mice. THE JOURNAL OF IMMUNOLOGY 2009; 182:5960-9. [PMID: 19414747 DOI: 10.4049/jimmunol.0900008] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
Recombinant lentivector immunization has been demonstrated to induce potent CD8 T cell responses in vivo. In this study, we investigated whether lentivector delivering a self/tumor Ag, tyrosinase related protein 1 (TRP1), could stimulate effective antitumor T cell responses. We found that immunization with lentivector expressing mutated TRP1 Ag elicited potent CD8 T cell responses against multiple TRP1 epitopes. Importantly, the activated CD8 T cells effectively recognize wild-type TRP1 epitopes. At peak times, as many as 10% of CD8 T cells were effector cells against TRP1 Ag. These cells killed wild-type TRP1 peptide-pulsed target cells in vivo and produced IFN-gamma after ex vivo stimulation. The CD8 T cell responses were long-lasting (3-4 wk). Immunized mice were protected from B16 tumor cell challenge. In a therapeutic setting, lentivector immunization induced potent CD8 T cell responses in tumor bearing mice. The number of infiltrating T cells and the ratio of CD8/CD4 were dramatically increased in the tumors of immunized mice. The tumor-infiltrating CD8 T cells were functional and produced IFN-gamma. The potent CD8 T cell responses stimulated by lentivector immunization eliminated small 3-day s.c. B16 tumors and strongly inhibited the growth of more established 5-day tumors. These studies demonstrate that genetic immunization with lentivector expressing mutated self/tumor Ag can generate potent CD8 T cell immune responses and antitumor immunity that prevent and inhibit B16 tumor growth, suggesting that lentivector immunization has the potential for tumor immunotherapy and immune prevention.
Collapse
Affiliation(s)
- Yanjun Liu
- Immunology/Immunotherapy Program, Medical College of Georgia Cancer Center, Medical College of Georgia, Augusta, GA 30912, USA
| | | | | | | | | | | | | |
Collapse
|
55
|
Kilinc MO, Gu T, Harden JL, Virtuoso LP, Egilmez NK. Central role of tumor-associated CD8+ T effector/memory cells in restoring systemic antitumor immunity. THE JOURNAL OF IMMUNOLOGY 2009; 182:4217-25. [PMID: 19299720 DOI: 10.4049/jimmunol.0802793] [Citation(s) in RCA: 44] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
Sustained delivery of IL-12 and GM-CSF to tumors induces the activation of tumor-resident CD8(+) T effector/memory cells (Tem) followed by cytotoxic CD8(+) T effector cell expansion. To determine whether the secondary effectors expanded from tumor-associated Tem or were primed de novo, activation kinetics of tumor-draining lymph node (TDLN) CD8(+) T cells were analyzed. Treatment promoted a 4-fold increase in the numbers of TDLN CD8(+) T cells displaying a CD69(+)CCR5(+)CD62L(-) periphery-homing effector phenotype by day 4 posttherapy. Pulse labeling of tumor and TDLN T cells with BrdU confirmed that proliferation occurred exclusively within the draining lymph nodes between days 1 and 4 with subsequent migration of primed CD8(+) T effectors to tumors on day 7. Day 4 CD8(+) T effector cells preferentially homed to and lysed experimental, but not control, tumors, establishing tumor specificity. To determine whether the secondary CD8(+) T effector cell response was dependent on activation of tumor-resident CD8(+) Tem, mice that were selectively depleted of tumor-infiltrating CD8(+) T cells were treated and monitored for T effector priming. In the absence of tumor-resident CD8(+) Tem, T effector cell expansion was completely abrogated in the TDLN, revealing that restoration of CD8(+) Tem function was critical to the induction of secondary T effectors. T cell priming failed to occur in IFN-gamma or perforin knockout mice, demonstrating that the requirement for Tem activation was associated with induction of Tem cytotoxicity. These data confirm that intratumoral IL-12 plus GM-CSF induces de novo priming of tumor-specific CD8(+) T effector cells in the TDLN and establish the critical role of preexisting intratumoral CD8(+) Tem in driving this process.
Collapse
Affiliation(s)
- Mehmet O Kilinc
- Department of Microbiology and Immunology, University at Buffalo, State University of New York, 14214, USA
| | | | | | | | | |
Collapse
|
56
|
Abstract
The development of effective immunotherapy strategies for glioma requires adequate understanding of the unique immunological microenvironment in the central nervous system (CNS) and CNS tumors. Although the CNS is often considered to be an immunologically privileged site and poses unique challenges for the delivery of effector cells and molecules, recent advances in technology and discoveries in CNS immunology suggest novel mechanisms that may significantly improve the efficacy of immunotherapy against gliomas. In this review, we first summarize recent advances in the CNS and CNS tumor immunology. We address factors that may promote immune escape of gliomas. We also review advances in passive and active immunotherapy strategies for glioma, with an emphasis on lessons learned from recent early-phase clinical trials. We also discuss novel immunotherapy strategies that have been recently tested in non-CNS tumors and show great potential for application to gliomas. Finally, we discuss how each of these promising strategies can be combined to achieve clinical benefit for patients with gliomas.
Collapse
Affiliation(s)
- Hideho Okada
- Brain Tumor Program, University of Pittsburgh Cancer Institute, Pittsburgh, PA 15213, USA.
| | | | | | | | | | | | | |
Collapse
|
57
|
Abstract
Cancer immunotherapy seeks to mobilize a patient's immune system for therapeutic benefit. It can be passive, that is, transfer of immune effector cells (T cells) or proteins (antibodies), or active, that is, vaccination. Early clinical trials testing vaccination with ex vivo generated dendritic cells (DCs) pulsed with tumor antigens provide a proof-of-principle that therapeutic immunity can be elicited. Yet, the clinical benefit measured by regression of established tumors in patients with stage IV cancer has been observed in a fraction of patients only. The next generation of DC vaccines is expected to generate large numbers of high avidity effector CD8 T cells and to overcome regulatory T cells and suppressive environment established by tumors, a major obstacle in metastatic disease. Therapeutic vaccination protocols will combine improved DC vaccines with chemotherapy to exploit immunogenic chemotherapy regimens. We foresee adjuvant vaccination in patients with resected tumors but at high risk of relapse to be based on in vivo targeting of DCs with fusion proteins containing anti-DCs antibodies, antigens from tumor stem/propagating cells, and DC activators.
Collapse
|
58
|
Abstract
The role that the immune system plays in limiting tumor formation and growth is becoming increasingly clear and passive immunotherapeutic approaches, such as the use of monoclonal antibodies, are now being successfully applied in clinical practice. Active immunization against tumors, however, has not yet been shown to have the same level of clinical efficacy. Two important reasons for this lack of efficacy have to do with the antigens being targeted, as well as the immunization approaches that have been tested. This review will highlight some of the requirements thought to be important for the successful development of an active immunization approach, with a focus on the ongoing development efforts for a novel agent targeting the cytochrome P450 family member, CYP1B1.
Collapse
Affiliation(s)
- Thomas M Luby
- MGI Pharma, 35 Hartwell Avenue, Lexington, MA 02421, USA.
| |
Collapse
|
59
|
Ahmadi M, Emery DC, Morgan DJ. Prevention of both direct and cross-priming of antitumor CD8+ T-cell responses following overproduction of prostaglandin E2 by tumor cells in vivo. Cancer Res 2008; 68:7520-9. [PMID: 18794140 DOI: 10.1158/0008-5472.can-08-1060] [Citation(s) in RCA: 81] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Defects in antitumor immune responses have been associated with increased release of prostaglandin E(2) (PGE(2)) as a result of overexpression of cyclooxygenase (COX)-2 by tumors. In this report, we examine the effects of PGE(2) on antitumor CD8(+) T-cell responses generated both by cross-presenting dendritic cells and by direct priming by tumor cells. Our data show that PGE(2) inhibits dendritic cell maturation, resulting in the abortive activation of naive CD8(+) T cells, and is dependent on interleukin-10 production by dendritic cells. Interaction of tumor cells with naïve CD8(+) T cells in the presence of PGE(2) in vitro results in the induction of CD8(+) CD28(-) T cells, which fail to proliferate or exhibit effector function. In vivo, overexpression of COX-2 by tumor cells results in a decrease in number of tumor-infiltrating dendritic cells and confers the ability of tumor cells to metastasize to the tumor draining lymph nodes.
Collapse
Affiliation(s)
- Maryam Ahmadi
- Department of Cellular and Molecular Medicine, University of Bristol, School of Medical Sciences, UK
| | | | | |
Collapse
|