1
|
Lv K, Yin T, Yu M, Chen Z, Zhou Y, Li F. Treatment Advances in EBV Related Lymphoproliferative Diseases. Front Oncol 2022; 12:838817. [PMID: 35515118 PMCID: PMC9063483 DOI: 10.3389/fonc.2022.838817] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2021] [Accepted: 03/11/2022] [Indexed: 12/24/2022] Open
Abstract
Epstein Barr virus (EBV) can affect 90% of the human population. It can invade B lymphocytes, T lymphocytes and natural killer cells of the host and remain in the host for life. The long latency and reactivation of EBV can cause malignant transformation, leading to various lymphoproliferative diseases (LPDs), including EBV-related B-cell lymphoproliferative diseases (EBV-B-LPDs) (for example, Burkitt lymphoma (BL), classic Hodgkin's lymphoma (cHL), and posttransplantation and HIV-related lymphoproliferative diseases) and EBV-related T-cell lymphoproliferative diseases (EBV-T/NK-LPDs) (for example, extranodal nasal type natural killer/T-cell lymphoma (ENKTCL), aggressive NK cell leukaemia (ANKL), and peripheral T-cell lymphoma, not otherwise specified (PTCL-NOS). EBV-LPDs are heterogeneous with different clinical features and prognoses. The treatment of EBV-LPDs is usually similar to that of EBV-negative lymphoma with the same histology and can include chemotherapy, radiotherapy, and hematopoietic stem cell transplant (HSCT). However, problems such as serious toxicity and drug resistance worsen the survival prognosis of patients. EBV expresses a variety of viral and lytic proteins that regulate cell cycle and death processes and promote the survival of tumour cells. Based on these characteristics, a series of treatment strategies for EBV in related malignant tumours have been developed, such as monoclonal antibodies, immune checkpoint inhibitors, cytotoxic T lymphocytes (CTLs) and epigenetic therapy. These new individualized therapies can produce highly specific killing effects on tumour cells, and nontumour cells can be protected from toxicity. This paper will focus on the latest progress in the treatment of EBV-LPDs based on pathological mechanisms.
Collapse
Affiliation(s)
- Kebing Lv
- Center of Hematology, The First Affiliated Hospital of Nanchang University, Nanchang, China
| | - Ting Yin
- Center of Hematology, The First Affiliated Hospital of Nanchang University, Nanchang, China
| | - Min Yu
- Center of Hematology, The First Affiliated Hospital of Nanchang University, Nanchang, China.,Institute of Hematology, Academy of Clinical Medicine of Jiangxi Province, Nanchang, China.,Clinical Research Center for Hematologic Disease of Jiangxi Province, Nanchang, China.,Institute of Lymphoma and Myeloma, Nanchang University, Nanchang, China
| | - Zhiwei Chen
- Center of Hematology, The First Affiliated Hospital of Nanchang University, Nanchang, China.,Institute of Hematology, Academy of Clinical Medicine of Jiangxi Province, Nanchang, China.,Clinical Research Center for Hematologic Disease of Jiangxi Province, Nanchang, China.,Institute of Lymphoma and Myeloma, Nanchang University, Nanchang, China
| | - Yulan Zhou
- Center of Hematology, The First Affiliated Hospital of Nanchang University, Nanchang, China.,Institute of Hematology, Academy of Clinical Medicine of Jiangxi Province, Nanchang, China.,Clinical Research Center for Hematologic Disease of Jiangxi Province, Nanchang, China.,Institute of Lymphoma and Myeloma, Nanchang University, Nanchang, China
| | - Fei Li
- Center of Hematology, The First Affiliated Hospital of Nanchang University, Nanchang, China.,Institute of Hematology, Academy of Clinical Medicine of Jiangxi Province, Nanchang, China.,Clinical Research Center for Hematologic Disease of Jiangxi Province, Nanchang, China.,Institute of Lymphoma and Myeloma, Nanchang University, Nanchang, China
| |
Collapse
|
2
|
Hammink R, Weiden J, Voerman D, Popelier C, Eggermont LJ, Schluck M, Figdor CG, Verdoes M. Semiflexible Immunobrushes Induce Enhanced T Cell Activation and Expansion. ACS APPLIED MATERIALS & INTERFACES 2021; 13:16007-16018. [PMID: 33797875 PMCID: PMC8045021 DOI: 10.1021/acsami.0c21994] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/06/2023]
Abstract
A variety of bioactive materials developed to expand T cells for adoptive transfer into cancer patients are currently evaluated in the clinic. In most cases, T cell activating biomolecules are attached to rigid surfaces or matrices and form a static interface between materials and the signaling receptors on the T cells. We hypothesized that a T cell activating polymer brush interface might better mimic the cell surface of a natural antigen-presenting cell, facilitating receptor movement and concomitant advantageous mechanical forces to provide enhanced T cell activating capacities. Here, as a proof of concept, we synthesized semiflexible polyisocyanopeptide (PIC) polymer-based immunobrushes equipped with T cell activating agonistic anti-CD3 (αCD3) and αCD28 antibodies placed on magnetic microbeads. We demonstrated enhanced efficiency of ex vivo expansion of activated primary human T cells even at very low numbers of stimulating antibodies compared to rigid beads. Importantly, the immunobrush architecture appeared crucial for this improved T cell activating capacity. Immunobrushes outperform current benchmarks by producing higher numbers of T cells exhibiting a combination of beneficial phenotypic characteristics, such as reduced exhaustion marker expression, high cytokine production, and robust expression of cytotoxic hallmarks. This study indicates that semiflexible immunobrushes have great potential in making T cell-based immunotherapies more effective.
Collapse
Affiliation(s)
- Roel Hammink
- Department
of Tumor Immunology, Radboud Institute for Molecular Life Sciences, Radboud University Medical Center, Geert Grooteplein 26, 6525 GA Nijmegen, Netherlands
- Division
of Immunotherapy, Oncode Institute, Radboud
University Medical Center, 6525 GA Nijmegen, Netherlands
| | - Jorieke Weiden
- Department
of Tumor Immunology, Radboud Institute for Molecular Life Sciences, Radboud University Medical Center, Geert Grooteplein 26, 6525 GA Nijmegen, Netherlands
- Division
of Immunotherapy, Oncode Institute, Radboud
University Medical Center, 6525 GA Nijmegen, Netherlands
- Institute
for Chemical Immunology, 6525 GA Nijmegen, Netherlands
| | - Dion Voerman
- Department
of Tumor Immunology, Radboud Institute for Molecular Life Sciences, Radboud University Medical Center, Geert Grooteplein 26, 6525 GA Nijmegen, Netherlands
- Institute
for Chemical Immunology, 6525 GA Nijmegen, Netherlands
| | - Carlijn Popelier
- Department
of Tumor Immunology, Radboud Institute for Molecular Life Sciences, Radboud University Medical Center, Geert Grooteplein 26, 6525 GA Nijmegen, Netherlands
| | - Loek J. Eggermont
- Department
of Tumor Immunology, Radboud Institute for Molecular Life Sciences, Radboud University Medical Center, Geert Grooteplein 26, 6525 GA Nijmegen, Netherlands
- Institute
for Chemical Immunology, 6525 GA Nijmegen, Netherlands
| | - Marjolein Schluck
- Department
of Tumor Immunology, Radboud Institute for Molecular Life Sciences, Radboud University Medical Center, Geert Grooteplein 26, 6525 GA Nijmegen, Netherlands
- Division
of Immunotherapy, Oncode Institute, Radboud
University Medical Center, 6525 GA Nijmegen, Netherlands
- Institute
for Chemical Immunology, 6525 GA Nijmegen, Netherlands
| | - Carl G. Figdor
- Department
of Tumor Immunology, Radboud Institute for Molecular Life Sciences, Radboud University Medical Center, Geert Grooteplein 26, 6525 GA Nijmegen, Netherlands
- Division
of Immunotherapy, Oncode Institute, Radboud
University Medical Center, 6525 GA Nijmegen, Netherlands
- Institute
for Chemical Immunology, 6525 GA Nijmegen, Netherlands
| | - Martijn Verdoes
- Department
of Tumor Immunology, Radboud Institute for Molecular Life Sciences, Radboud University Medical Center, Geert Grooteplein 26, 6525 GA Nijmegen, Netherlands
- Institute
for Chemical Immunology, 6525 GA Nijmegen, Netherlands
| |
Collapse
|
3
|
Muhammad Q, Jang Y, Kang SH, Moon J, Kim WJ, Park H. Modulation of immune responses with nanoparticles and reduction of their immunotoxicity. Biomater Sci 2020; 8:1490-1501. [PMID: 31994542 DOI: 10.1039/c9bm01643k] [Citation(s) in RCA: 35] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Particles with a size range of 1-100 nm used in various fields of life sciences are called nanoparticles (NPs). Currently, nanotechnology has a wide range of applications in biomedical research, industries and in almost all types of modern technology. The growing applications of nanotechnology in medicine urge scientists to analyze the impact of NPs on human body tissues and the immune system. Easy surface modifications of the NPs enable the modulation of the immune system either by evading the immune system to prevent allergic reactions or by enhancing the immunogenic response. In this review, we discussed the various possible theories and practical implications reported to date for the applications of nanotechnology in immunostimulation and immunosuppression for favorable immune response, such as vaccine delivery and cancer treatments. In the last part of this paper, we also discussed the biocompatibility and unfavorable immunotoxicity of NPs and methods for lowering their toxicity.
Collapse
Affiliation(s)
- Qasim Muhammad
- School of Integrative Engineering, Chung-Ang University, 221 Heukseok-Dong, Dongjak-Gu, Seoul 06974, Republic of Korea.
| | - Yeonwoo Jang
- School of Integrative Engineering, Chung-Ang University, 221 Heukseok-Dong, Dongjak-Gu, Seoul 06974, Republic of Korea.
| | - Shin Hyuk Kang
- Department of Plastic and Reconstructive Surgery, Seoul National University Bundang Hospital, Seongnam 13620, Republic of Korea
| | - James Moon
- Pharmaceutical Sciences and Biomedical Engineering, University of Michigan, 500 S. State Street, Ann Arbor, MI 48109, USA
| | - Won Jong Kim
- School of Interdisciplinary Bioscience and Bioengineering, Pohang University of Science and Technology (POSTECH), Pohang 37673, Republic of Korea.
| | - Hansoo Park
- School of Integrative Engineering, Chung-Ang University, 221 Heukseok-Dong, Dongjak-Gu, Seoul 06974, Republic of Korea.
| |
Collapse
|
4
|
Downey RF, Sullivan FJ, Wang-Johanning F, Ambs S, Giles FJ, Glynn SA. Human endogenous retrovirus K and cancer: Innocent bystander or tumorigenic accomplice? Int J Cancer 2014; 137:1249-57. [PMID: 24890612 PMCID: PMC6264888 DOI: 10.1002/ijc.29003] [Citation(s) in RCA: 93] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2013] [Revised: 05/02/2014] [Accepted: 05/09/2014] [Indexed: 12/20/2022]
Abstract
Harbored as relics of ancient germline infections, human endogenous retroviruses (HERVs) now constitute up to 8% of our genome. A proportion of this sequence has been co-opted for molecular and cellular processes, beneficial to human physiology, such as the fusogenic activity of the envelope protein, a vital component of placentogenesis. However, the discovery of high levels of HERV-K mRNA and protein and even virions in a wide array of cancers has revealed that HERV-K may be playing a more sinister role–a role as an etiological agent in cancer itself. Whether the presence of this retroviral material is simply an epiphenomenon, or an actual causative factor, is a hotly debated topic. This review will summarize the current state of knowledge regarding HERV-K and cancer and attempt to outline the potential mechanisms by which HERV-K could be involved in the onset and promotion of carcinogenesis.
Collapse
Affiliation(s)
- Ronan F Downey
- Prostate Cancer Institute, National University of Ireland Galway, Galway, Ireland
| | - Francis J Sullivan
- Prostate Cancer Institute, National University of Ireland Galway, Galway, Ireland.,Department of Radiation Oncology, Galway University Hospitals, Galway, Ireland
| | | | - Stefan Ambs
- Laboratory of Human Carcinogenesis, National Cancer Institute, Bethesda, MD
| | - Francis J Giles
- Prostate Cancer Institute, National University of Ireland Galway, Galway, Ireland.,HRB Clinical Research Facilities Galway & Dublin, National University of Ireland Galway and Trinity College Dublin, Galway, Ireland
| | - Sharon A Glynn
- Prostate Cancer Institute, National University of Ireland Galway, Galway, Ireland
| |
Collapse
|
5
|
Mautner J, Bornkamm GW. The role of virus-specific CD4+ T cells in the control of Epstein-Barr virus infection. Eur J Cell Biol 2011; 91:31-5. [PMID: 21458882 DOI: 10.1016/j.ejcb.2011.01.007] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2010] [Accepted: 01/22/2011] [Indexed: 11/15/2022] Open
Abstract
Epstein-Barr virus (EBV) establishes lifelong persistent infections in humans and has been implicated in the pathogenesis of several human malignancies. Protective immunity against EBV is mediated by T cells, as indicated by an increased incidence of EBV-associated malignancies in immunocompromised patients, and by the successful treatment of EBV-associated post-transplant lymphoproliferative disease (PTLD) in transplant recipients by the infusion of polyclonal EBV-specific T cell lines. To implement this treatment modality as a conventional therapeutic option, and to extend this protocol to other EBV-associated diseases, generic and more direct approaches for the generation of EBV-specific T cell lines enriched in disease-relevant specificities need to be developed. To this aim, we studied the poorly defined EBV-specific CD4+ T cell response during acute and chronic infection.
Collapse
Affiliation(s)
- Josef Mautner
- Clinical Cooperation Group, Pediatric Tumor Immunology, Helmholtz-Zentrum München, Marchioninistrasse 25, Munich, Germany.
| | | |
Collapse
|
6
|
Redirecting T-cell specificity by introducing a tumor-specific chimeric antigen receptor. Blood 2010; 116:1035-44. [PMID: 20439624 DOI: 10.1182/blood-2010-01-043737] [Citation(s) in RCA: 224] [Impact Index Per Article: 14.9] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Infusions of antigen-specific T cells have yielded therapeutic responses in patients with pathogens and tumors. To broaden the clinical application of adoptive immunotherapy against malignancies, investigators have developed robust systems for the genetic modification and characterization of T cells expressing introduced chimeric antigen receptors (CARs) to redirect specificity. Human trials are under way in patients with aggressive malignancies to test the hypothesis that manipulating the recipient and reprogramming T cells before adoptive transfer may improve their therapeutic effect. These examples of personalized medicine infuse T cells designed to meet patients' needs by redirecting their specificity to target molecular determinants on the underlying malignancy. The generation of clinical grade CAR(+) T cells is an example of bench-to-bedside translational science that has been accomplished using investigator-initiated trials operating largely without industry support. The next-generation trials will deliver designer T cells with improved homing, CAR-mediated signaling, and replicative potential, as investigators move from the bedside to the bench and back again.
Collapse
|
7
|
Sengar RS, Spokauskiene L, Steed DP, Griffin P, Arbujas N, Chambers WH, Wiener EC. Magnetic resonance imaging-guided adoptive cellular immunotherapy of central nervous system tumors with a T1 contrast agent. Magn Reson Med 2009; 62:599-606. [PMID: 19544372 DOI: 10.1002/mrm.22030] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/27/2023]
Abstract
Dendritic cells (DCs) are the most effective antigen-presenting cells (APCs) and are used in a variety of immunotherapeutic approaches. Adoptive cellular immunotherapy (ACI) of cancer using DCs has attracted much interest due to their capacity to promote immunity in prophylactic and therapeutic protocols. As one approach, DCs are injected into patients or tumor-bearing animals, to trigger specific antitumor immunity. In that framework, several approaches to DC delivery have been reported, including direct intratumoral injection; this has yielded positive but variable results. The underlying reasons for this have not been fully determined, but major hypotheses include technical difficulties in delivering cells into tumors and tumor-mediated immunosuppression. Image-guided ACI offers the potential to establish that DCs are efficiently delivered to the tumor site, which might eliminate some of the variability. Therefore, we developed highly sensitive methods for monitoring the injection or trafficking of DCs into tumors using a clinically approved formulation of a gadolinium-based magnetic resonance imaging (MRI) contrast agent, Gd(III)-HP-DO3A (ProHance). We determined the labeling efficiency of DCs with this formulation; that labeling DCs with this agent did not inhibit expression of surface markers important for antigen presentation and activation of naive T cells; that their capacity to interact with natural killer (NK) cells was not reduced; and that their migration was not diminished. Further, we determined that ProHance-labeled DCs can be effectively imaged in vivo in established central nervous system tumors.
Collapse
Affiliation(s)
- Raghvendra S Sengar
- Department of Radiology, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA
| | | | | | | | | | | | | |
Collapse
|
8
|
Wang H, Wei H, Zhang R, Hou S, Li B, Qian W, Zhang D, Kou G, Dai J, Guo Y. Genetically targeted T cells eradicate established breast cancer in syngeneic mice. Clin Cancer Res 2009; 15:943-50. [PMID: 19188165 DOI: 10.1158/1078-0432.ccr-08-2381] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
PURPOSE The purpose of the present study was to evaluate the capacity and mechanisms of genetically modified erbB2-specific T cells to eradicate erbB2+ tumors in syngeneic mice. EXPERIMENTAL DESIGN Primary mouse T cells were modified to target the breast tumor-associated antigen erbB2 through retroviral-mediated transfer of a chimeric antigen receptor, termed single-chain antibody (scFv)-CD28-zeta. Antitumor efficacy of scFv-CD28-zeta-modified T cells was analyzed in mice bearing D2F2/E2 breast tumors. RESULTS The scFv-CD28-zeta-modified T cells were shown to specifically secrete T cytotoxic-1 cytokines and lyse erbB2+ breast tumor cells following receptor stimulation in vitro. Treatment with scFv-CD28-zeta-modified T cells was able to lead to long-term, tumor-free survival in mice bearing erbB2+ D2F2/E2 breast tumors. Importantly, the surviving mice developed a host memory response to D2F2/E2 tumor cells, and this host response was able to protect against a rechallenge with erbB2+ D2F2/E2 tumor cells and parental erbB2(-) D2F2 tumor cells. In addition, scFv-CD28-zeta T-cell expression of perforin and interferon-gamma were essential for complete antitumor efficacy. CONCLUSIONS Treatment with scFv-CD28-zeta-modified T cells was able to induce a host antitumor immunity in syngeneic mice. Complete tumor elimination by scFv-CD28-zeta-modified T cells required T cell-derived interferon-gamma and perforin, indicating that cytotoxicity and cytokine secretion play a role in the in vivo response.
Collapse
Affiliation(s)
- Hao Wang
- International Joint Cancer Institute and Changhai Hospital Cancer Center, The Second Military Medical University, Shanghai, People's Republic of China
| | | | | | | | | | | | | | | | | | | |
Collapse
|
9
|
Ando T, Mimura K, Johansson CC, Hanson MG, Mougiakakos D, Larsson C, Martins da Palma T, Sakurai D, Norell H, Li M, Nishimura MI, Kiessling R. Transduction with the antioxidant enzyme catalase protects human T cells against oxidative stress. THE JOURNAL OF IMMUNOLOGY 2009; 181:8382-90. [PMID: 19050255 DOI: 10.4049/jimmunol.181.12.8382] [Citation(s) in RCA: 74] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
Abstract
Patients with diseases characterized by chronic inflammation, caused by infection or cancer, have T cells and NK cells with impaired function. The underlying molecular mechanisms are diverse, but one of the major mediators in this immune suppression is oxidative stress caused by activated monocytes, granulocytes, or myeloid-derived suppressor cells. Reactive oxygen species can seriously hamper the efficacy of active immunotherapy and adoptive transfer of T and NK cells into patients. In this study, we have evaluated whether enhanced expression of the antioxidant enzyme catalase in human T cells can protect them against reactive oxygen species. Human CD4(+) and CD8(+) T cells retrovirally transduced with the catalase gene had increased intracellular expression and activity of catalase. Catalase transduction made CD4(+) T cells less sensitive to H(2)O(2)-induced loss-of-function, measured by their cytokine production and ability to expand in vitro following anti-CD3 stimulation. It also enhanced the resistance to oxidative stress-induced cell death after coculture with activated granulocytes, exposure to the oxidized lipid 4-hydroxynonenal, or H(2)O(2). Expression of catalase by CMV-specific CD8(+) T cells saved cells from cell death and improved their capacity to recognize CMV peptide-loaded target cells when exposed to H(2)O(2). These findings indicate that catalase-transduced T cells potentially are more efficacious for the immunotherapy of patients with advanced cancer or chronic viral infections.
Collapse
Affiliation(s)
- Takashi Ando
- Department of Oncology and Pathology, Immune and Gene Therapy Laboratory, Cancer Center Karolinska, Karolinska Institutet, Stockholm, Sweden
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
10
|
Wang-Johanning F, Radvanyi L, Rycaj K, Plummer JB, Yan P, Sastry KJ, Piyathilake CJ, Hunt KK, Johanning GL. Human endogenous retrovirus K triggers an antigen-specific immune response in breast cancer patients. Cancer Res 2008; 68:5869-77. [PMID: 18632641 DOI: 10.1158/0008-5472.can-07-6838] [Citation(s) in RCA: 148] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
Recent evidence indicates that human cancer cells reactivate the expression of latent human endogenous retroviral (HERV) proteins. However, the extent to which cancer patients mount de novo immune responses against expressed HERV elements is unclear. In this study, we determined the extent of HERV-K env expression in human breast cancer (BC) and whether both humoral and cell-mediated immunity against HERV-K can be found in BC patients. We found HERV-K env protein expression in 88% of BC (n = 119) but not in normal breast (n = 76) tissues. ELISA screening assays detected significant titers of anti-HERV-K env IgG in a large proportion of BC patients. T-cell responses against HERV-K were also detected in peripheral blood mononuclear cells (PBMC) from BC patients stimulated with autologous dendritic cells pulsed with HERV-K env SU antigens. These responses included induction of T-cell proliferation (P = 0.0043), IFN-gamma production measured by enzyme-linked immunospot (P < 0.0001), and multiplex cytokine secretion (P = 0.0033). Multiplex cytokine analysis found a T-helper 1 cytokine response, including interleukin (IL)-2 (P = 0.0109), IL-6 (P = 0.0396), IL-8 (P = 0.0169), and IP-10 (P = 0.0045) secretion during in vitro stimulation of BC PBMC with HERV-K antigen. We also found HERV-K-specific CTLs that were capable of lysing target cells expressing HERV-K env protein in BC patients but not in normal female controls without cancer. These findings suggest that retroviral gene products are capable of acting as tumor-associated antigens activating both T-cell and B-cell responses in BC patients.
Collapse
Affiliation(s)
- Feng Wang-Johanning
- Department of Veterinary Sciences and Michale E. Keeling Center for Comparative Medicine and Research, The University of Texas M. D. Anderson Cancer Center, Houston, TX 78602-6621, USA.
| | | | | | | | | | | | | | | | | |
Collapse
|
11
|
Abstract
Clinical trials have established that T cells have the ability to prevent and treat pathogens and tumors. This is perhaps best exemplified by engraftment of allogeneic T cells in the context of hematopoietic stem-cell transplantation (HSCT), which for over the last 50 years remains one of the best and most robust examples of cell-based therapies for the treatment of hematologic malignancies. Yet, the approach to infuse T cells for treatment of cancer, in general, and pediatric tumors, in particular, generally remains on the sidelines of cancer therapy. This review outlines the current state-of-the-art and provides a rationale for undertaking adoptive immunotherapy trials with emphasis on childhood malignancies.
Collapse
|
12
|
Autologous white blood cell transfusion: Toward a younger immunity. Hum Immunol 2007; 68:805-12. [DOI: 10.1016/j.humimm.2007.07.004] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2007] [Revised: 07/05/2007] [Accepted: 07/16/2007] [Indexed: 12/31/2022]
|
13
|
Savai R, Schermuly RT, Pullamsetti SS, Schneider M, Greschus S, Ghofrani HA, Traupe H, Grimminger F, Banat GA. A combination hybrid-based vaccination/adoptive cellular therapy to prevent tumor growth by involvement of T cells. Cancer Res 2007; 67:5443-53. [PMID: 17545626 DOI: 10.1158/0008-5472.can-06-3677] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Cancer immunotherapy with dendritic cell-tumor cell fusion hybrids induces polyclonal stimulation against a variety of tumor antigens, including unknown antigens. Hybrid cells can prime CTLs, which subsequently develop antitumor responses. The aim of this study was to enhance the known antitumor effect of hybrid vaccination (HC-Vacc) and hybrid-primed adoptive T-cell therapy (HC-ACT) using the poorly immunogenic Lewis lung carcinoma (LLC1) model. The strategy used was a combination of a double HC-Vacc alternating with HC-ACT (HC-Vacc/ACT). Using flat-panel volumetric computer tomography and immunohistochemistry, we showed a significant retardation of tumor growth (85%). In addition, a significant delay in tumor development, a reduction in the number of pulmonary metastases, and increased survival times were observed. Furthermore, the tumors displayed significant morphologic changes and increased apoptosis, as shown by up-regulation of gene expression of the proapoptotic markers Fas, caspase-8, and caspase-3. The residual tumor masses seen in the HC-Vacc/ACT-treated mice were infiltrated with CD4+ and CD8+ lymphocytes and showed elevated IFNgamma expression. Moreover, splenic enlargement observed in HC-Vacc/ACT-treated mice reflected the increased functionality of T cells, as also indicated by increased expression of markers for CTL activation, differentiation, and proliferation (Cd28, Icosl, Tnfrsf13, and Tnfsf14). Our findings indicate that the combination therapy of dendritic cell-tumor cell HC-Vacc/ACT is a very effective and a promising immunotherapeutic regimen against poorly immunogenic carcinomas.
Collapse
MESH Headings
- Animals
- Apoptosis/genetics
- Cancer Vaccines/immunology
- Cancer Vaccines/pharmacology
- Carcinoma, Lewis Lung/genetics
- Carcinoma, Lewis Lung/immunology
- Carcinoma, Lewis Lung/pathology
- Carcinoma, Lewis Lung/therapy
- Cell Fusion/methods
- Dendritic Cells/cytology
- Dendritic Cells/immunology
- Female
- Gene Expression
- Hybrid Cells/immunology
- Immunotherapy, Adoptive/methods
- Lung Neoplasms/secondary
- Lymphocyte Activation
- Melanoma, Experimental/genetics
- Melanoma, Experimental/immunology
- Melanoma, Experimental/pathology
- Melanoma, Experimental/therapy
- Mice
- Mice, Inbred BALB C
- Mice, Inbred C57BL
- T-Lymphocytes/immunology
- T-Lymphocytes, Cytotoxic/immunology
Collapse
Affiliation(s)
- Rajkumar Savai
- Department of Hematology, Universitätsklinikum Giessen und Marburg GmbH, Giessen, Germany
| | | | | | | | | | | | | | | | | |
Collapse
|
14
|
Adhikary D, Behrends U, Boerschmann H, Pfünder A, Burdach S, Moosmann A, Witter K, Bornkamm GW, Mautner J. Immunodominance of lytic cycle antigens in Epstein-Barr virus-specific CD4+ T cell preparations for therapy. PLoS One 2007; 2:e583. [PMID: 17611619 PMCID: PMC1894652 DOI: 10.1371/journal.pone.0000583] [Citation(s) in RCA: 61] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2007] [Accepted: 06/03/2007] [Indexed: 12/15/2022] Open
Abstract
BACKGROUND Epstein-Barr virus (EBV) is associated with a number of human malignancies. EBV-positive post-transplant lymphoproliferative disease in solid organ and hematopoietic stem cell transplant recipients has been successfully treated by the adoptive transfer of polyclonal EBV-specific T cell lines containing CD4+ and CD8+ T cell components. Although patients receiving T cell preparations with a higher CD4+ T cell proportion show better clinical responses, the specificity of the infused CD4+ component has remained completely unknown. METHODOLOGY/PRINCIPAL FINDINGS We generated LCL-stimulated T cell lines from 21 donors according to clinical protocols, and analyzed the antigen specificity of the CD4+ component in EBV-specific T cell preparations using a genetically engineered EBV mutant that is unable to enter the lytic cycle, and recombinantly expressed and purified EBV proteins. Surprisingly, CD4+ T cell lines from acutely and persistently EBV-infected donors consistently responded against EBV lytic cycle antigens and autoantigens, but barely against latent cycle antigens of EBV hitherto considered principal immunotherapeutic targets. Lytic cycle antigens were predominantly derived from structural proteins of the virus presented on MHC II via receptor-mediated uptake of released viral particles, but also included abundant infected cell proteins whose presentation involved intercellular protein transfer. Importantly, presentation of virion antigens was severely impaired by acyclovir treatment of stimulator cells, as currently performed in most clinical protocols. CONCLUSIONS/SIGNIFICANCE These results indicate that structural antigens of EBV are the immunodominant targets of CD4+ T cells in LCL-stimulated T cell preparations. These findings add to our understanding of the immune response against this human tumor-virus and have important implications for the improvement of immunotherapeutic strategies against EBV.
Collapse
Affiliation(s)
- Dinesh Adhikary
- Clinical Cooperation Group, Institute for Clinical and Molecular Biology, GSF-National Research Center for Environment and Health, Munich, Germany; Children's Hospital, Hematology-Oncology, University of Technology, Munich, Germany
| | | | | | | | | | | | | | | | | |
Collapse
|
15
|
Wrzesinski C, Paulos CM, Gattinoni L, Palmer DC, Kaiser A, Yu Z, Rosenberg SA, Restifo NP. Hematopoietic stem cells promote the expansion and function of adoptively transferred antitumor CD8 T cells. J Clin Invest 2007; 117:492-501. [PMID: 17273561 PMCID: PMC1783812 DOI: 10.1172/jci30414] [Citation(s) in RCA: 155] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2006] [Accepted: 11/21/2006] [Indexed: 01/22/2023] Open
Abstract
Depleting host immune elements with nonmyeloablative regimens prior to the adoptive transfer of tumor-specific CD8(+) T cells significantly enhances tumor treatment. In the current study, superior antitumor efficacy was achieved by further increasing the intensity of lymphodepletion to a level that required HSC transplantation. Surprisingly, the HSC transplant and not the increased lymphodepletion caused a robust expansion of adoptively transferred tumor-specific CD8(+) T cells. The HSC-driven cell expansion of effector, but not of naive, CD8(+) T cells was independent of in vivo restimulation by MHC class I-expressing APCs. Simultaneously, HSCs also facilitated the reconstitution of the host lymphoid compartment, including inhibitory elements, not merely via the production of progeny cells but by enhancing the expansion of cells that had survived lymphodepletion. Profound lymphodepletion, by myeloablation or by genetic means, focused the nonspecific HSC boost preferentially toward the transferred tumor-specific T cells, leading to successful tumor treatment. These findings indicate that CD8(+) T cell-mediated tumor responses can be efficiently driven by HSCs in the myeloablative setting and have substantial implications for the design of new antitumor immunotherapies.
Collapse
|
16
|
Abstract
Epstein-Barr virus (EBV), discovered > 40 years ago from a Burkitt's lymphoma biopsy, was the first virus to be directly associated with human cancer. EBV has two distinct life cycles in the human host; a lytic form of infection that produces new infectious virions, and a latent form of infection that allows the virus to persist in a dormant state for the lifetime of the host. EBV has evolved a life cycle that mimics the natural differentiation pathway of antigen-activated B cells, giving the virus access to its site of latent infection, the resting memory B cell. By steering infected cells through the various stages of lymphocyte differentiation, EBV is able to enter a cell type suitable for long-term latent persistence and periodic reactivation. However, its presence in various stages of B-cell development, and its ability to infect certain epithelial cells, can have pathogenic consequences, and can contribute to the development of a diverse group of lymphomas and carcinomas. The presence of EBV in the tumour cells of EBV-associated cancers might provide a basis for specific therapy. This article focuses on the contributions that the virus may play in different types of human cancer, particularly Burkitt's lymphoma, Hodgkin's lymphoma, lymphomas and lymphoproliferative diseases in the immunocompromised, and nasopharyngeal and gastric carcinoma.
Collapse
Affiliation(s)
- Samuel B Pattle
- Imperial College Faculty of Medicine, Department of Virology, Norfolk Place, London, W2 1PG, UK.
| | | |
Collapse
|