151
|
Abstract
Among the novel biological therapeutics that will increase our ability to cure human cancer in years to come, adoptive cellular therapy is one of the most promising approaches. Although this is a complex and challenging field, there have been major advances in basic and translational research resulting in clinical trial activity that is now beginning to confirm this promise. The results obtained with tumor-infiltrating lymphocytes therapy for melanoma, and virus-specific CTLs for EBV-associated malignancies are encouraging in terms of both ability to obtain clinical benefit and limited toxicity profile. In both settings, objective responses were obtained in at least 50% of treated patients. However, improvements to the clinical protocols, in terms of better patient selection and timing of administration, as well as cell product quality and availability, are clearly necessary to further ameliorate outcome, and logistical solutions are warranted to extend T-cell therapy beyond academic centers. In particular, there is a need to simplify cell production, in order to decrease costs and ease preparation. Promising implementations are underway, including harnessing the therapeutic potential of T cells transduced with TCRs directed against shared tumor antigens, and delineating strategies aimed at targeting immune evasion mechanisms exerted by tumor cells.
Collapse
|
152
|
Reconstituted complexes of mycobacterial HSP70 and EBV LMP2A-derived peptides elicit peptide-specific cytotoxic T lymphocyte responses and anti-tumor immunity. Vaccine 2011; 29:7414-23. [PMID: 21807054 DOI: 10.1016/j.vaccine.2011.07.063] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2011] [Revised: 06/30/2011] [Accepted: 07/17/2011] [Indexed: 11/20/2022]
Abstract
Epstein-Barr virus (EBV) latent membrane protein 2A (LMP2A) is a subdominant antigen expressed in EBV-associated malignancies, such as Hodgkin's diseases (HD) and nasopharyngeal carcinoma. A large number of previous studies have described LMP2A as an ideal target antigen in immunotherapy of EBV-related diseases, while limited successes have been achieved in clinical trials. Mycobacterium tuberculosis heat shock protein 70 (MtHsp70) is known as an effective molecular adjuvant for protein- or epitope-based vaccines. In the present study, we reconstituted two chaperone complexes of MtHsp70 and LMP2A-derived peptides (LMP2A(356-364) FLYALALLL and LMP2A(426-434) CLGGLLTMV) in vitro. We then investigated LMP2A-specific immune responses induced by reconstituted complexes of MtHsp70 and LMP2A-peptides using both EBV infected healthy donor PBMCs and HLA-A2.1 transgenic mouse models. We found that reconstituted complexes of MtHsp70 and LMP2A-peptides significantly elicit LMP2A-specific IFN-γ-producing cells and rousted cytotoxic T lymphocytes (CTLs) in vitro and in vivo. In addition, LMP2A-specific immune responses induced by the reconstituted complexes of MtHsp70 and LMP2A-peptides mediated potently protective activity as well as therapeutic efficacy against LMP2A-expressed tumor challenge in mouse models. These studies provide new insights for the development of novel LMP2A-based vaccines against EBV-associated malignancies.
Collapse
|
153
|
Rosenberg SA, Yang JC, Sherry RM, Kammula US, Hughes MS, Phan GQ, Citrin DE, Restifo NP, Robbins PF, Wunderlich JR, Morton KE, Laurencot CM, Steinberg SM, White DE, Dudley ME. Durable complete responses in heavily pretreated patients with metastatic melanoma using T-cell transfer immunotherapy. Clin Cancer Res 2011; 17:4550-7. [PMID: 21498393 PMCID: PMC3131487 DOI: 10.1158/1078-0432.ccr-11-0116] [Citation(s) in RCA: 1619] [Impact Index Per Article: 115.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023]
Abstract
PURPOSE Most treatments for patients with metastatic melanoma have a low rate of complete regression and thus overall survival in these patients is poor. We investigated the ability of adoptive cell transfer utilizing autologous tumor-infiltrating lymphocytes (TIL) to mediate durable complete regressions in heavily pretreated patients with metastatic melanoma. EXPERIMENTAL DESIGN Ninety-three patients with measurable metastatic melanoma were treated with the adoptive transfer of autologous TILs administered in conjunction with interleukin-2 following a lymphodepleting preparative regimen on three sequential clinical trials. Ninety-five percent of these patients had progressive disease following a prior systemic treatment. Median potential follow-up was 62 months. RESULTS Objective response rates by Response Evaluation Criteria in Solid Tumors (RECIST) in the 3 trials using lymphodepleting preparative regimens (chemotherapy alone or with 2 or 12 Gy irradiation) were 49%, 52%, and 72%, respectively. Twenty of the 93 patients (22%) achieved a complete tumor regression, and 19 have ongoing complete regressions beyond 3 years. The actuarial 3- and 5-year survival rates for the entire group were 36% and 29%, respectively, but for the 20 complete responders were 100% and 93%. The likelihood of achieving a complete response was similar regardless of prior therapy. Factors associated with objective response included longer telomeres of the infused cells, the number of CD8(+)CD27(+) cells infused, and the persistence of the infused cells in the circulation at 1 month (all P(2) < 0.001). CONCLUSIONS Cell transfer therapy with autologous TILs can mediate durable complete responses in patients with metastatic melanoma and has similar efficacy irrespective of prior treatment. Clin Cancer Res; 17(13); 4550-7. ©2011 AACR.
Collapse
Affiliation(s)
- Steven A Rosenberg
- Surgery Branch, National Cancer Institute, NIH, Bethesda, Maryland, USA.
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
154
|
Pedrazzoli P, Secondino S, Perfetti V, Comoli P, Montagna D. Immunotherapeutic Intervention against Sarcomas. J Cancer 2011; 2:350-6. [PMID: 21716856 PMCID: PMC3119402 DOI: 10.7150/jca.2.350] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2011] [Accepted: 06/03/2011] [Indexed: 12/16/2022] Open
Abstract
Advances in systemic therapy for sarcoma have produced, over the last two decades, relatively short-term benefits for the majority of patient. Among the novel biologic therapeutics that will likely increase our ability to cure human cancer in the years to come, immunotherapy is one of the most promising approaches. While past attempts to use immunotherapy have failed to dramatically shift the paradigm of care for the treatment of patients with sarcoma, major advances in basic and translational research have resulted, in more recent years, in clinical trial activity that is now beginning to generate promising results. However, to move from “proof of principle” to large scale clinical applicability, we need well-designed, multi-institutional clinical trials, along with continuous laboratory research to explore further the immunological characteristics of individual sarcoma subtypes and the consequent tailoring of therapy.
Collapse
Affiliation(s)
- Paolo Pedrazzoli
- 1. SC Oncologia, Fondazione IRCCS Policlinico S. Matteo, Pavia, Italy
| | | | | | | | | |
Collapse
|
155
|
Basso S, Zecca M, Merli P, Gurrado A, Secondino S, Quartuccio G, Guido I, Guerini P, Ottonello G, Zavras N, Maccario R, Pedrazzoli P, Comoli P. T cell therapy for nasopharyngeal carcinoma. J Cancer 2011; 2:341-6. [PMID: 21716854 PMCID: PMC3119400 DOI: 10.7150/jca.2.341] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2011] [Accepted: 06/02/2011] [Indexed: 01/29/2023] Open
Abstract
Among the novel biologic therapeutics that will increase our ability to cure human cancer in the years to come, T cell therapy is one of the most promising approaches. However, with the possible exception of tumor-infiltrating lymphocytes therapy for melanoma, clinical trials of adoptive T-cell therapy for solid tumors have so far provided only clear proofs-of-principle to build on with further development. Epstein-Barr virus (EBV)-associated malignancies offer a unique model to develop T cell-based immune therapies, targeting viral antigens expressed on tumor cells. In the last two decades, EBV-specific cytotoxic T-lymphocytes (CTL) have been successfully employed for the prophylaxis and treatment of EBV-related lymphoproliferative disorders in immunocompromised hosts. More recently, this therapeutic approach has been applied to the setting of EBV-related solid tumors, such as nasopharyngeal carcinoma. The results are encouraging, although further improvements to the clinical protocols are clearly necessary to increase anti-tumor activity. Promising implementations are underway, including harnessing the therapeutic potential of CTLs specific for subdominant EBV latent cycle epitopes, and delineating strategies aimed at targeting immune evasion mechanisms exerted by tumor cells.
Collapse
Affiliation(s)
- S Basso
- Pediatric Hematology/Oncology, Research Laboratories, and Medical Oncology, Fondazione IRCCS Policlinico S. Matteo, Pavia, Italy
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
156
|
Adoptive transfer of EBV-specific T cells results in sustained clinical responses in patients with locoregional nasopharyngeal carcinoma. J Immunother 2011; 33:983-90. [PMID: 20948438 DOI: 10.1097/cji.0b013e3181f3cbf4] [Citation(s) in RCA: 168] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Patients with recurrent or refractory Epstein Barr Virus (EBV)-positive nasopharyngeal carcinoma (NPC) continue to have poor outcomes. Our earlier Phase I dose escalation clinical study of 10 NPC patients showed that infusion of EBV-specific cytotoxic T cells (EBV-CTLs) was safe and had antitumor activity. To better define the overall response rate and discover whether disease status, EBV-antigen specificity, and/or in vivo expansion of infused EBV-CTLs predicted outcome, we treated 13 additional NPC patients with EBV-CTLs in a fixed-dose, Phase II component of the study. We assessed toxicity, efficacy, specificity, and expansion of infused CTLs for all 23 recurrent/refractory NPC patients treated on this Phase I/II clinical study. At the time of CTL infusion, 8 relapsed NPC patients were in remission and 15 had active disease. No significant toxicity was observed. Of the relapsed patients treated in their second or subsequent remission, 62% (5/8) remain disease free (at 17 to 75 mo), whereas 48.7% (7/15) of those with active disease had a CR/CRu (33.3%) or PR (15.4%). In contrast to locoregional disease, metastatic disease was associated with an increased risk of disease progression (HR: 3.91, P=0.015) and decreased overall survival (HR: 5.55, P=0.022). Neither the specificity of the infused CTLs for particular EBV antigens nor their measurable in vivo expansion discernibly influenced outcome. In conclusion, treatment of patients with relapsed/refractory EBV-positive NPC with EBV-CTLs is safe and can be associated with significant, long-term clinical benefit, particularly for patients with locoregional disease.
Collapse
|
157
|
Li J, Chen QY, Mo H, Zhang YL, Huang ZF, Zeng YX. Immunophenotyping at the time of diagnosis distinguishes two groups of nasopharyngeal carcinoma patients: implications for adoptive immunotherapy. Int J Biol Sci 2011; 7:607-17. [PMID: 21614153 PMCID: PMC3101529 DOI: 10.7150/ijbs.7.607] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2011] [Accepted: 05/08/2011] [Indexed: 02/05/2023] Open
Abstract
Background: Adoptive immunotherapy with EBV-specific CTLs (EBV-CTL) has been used to treat EBV-associated nasopharyngeal carcinoma (NPC) but only a fraction of the patients shows noticeable clinical response. Patients and Methods: Sixty-seven newly diagnosed NPC patients from 2005 to 2007 and 21 healthy donors were collected. Immunological parameters and immune function of PBMCs and EBV-CTL were analyzed by flow cytometer analysis (FACS) and 51Cr releasing experiment; Molecular characteristics on NPC tumor cells were investigated by immunochemical staining and statistic analysis. Results: NPC patients can be classified into two groups based on the percentage of CD3+ T cells in peripheral blood before accepted any treatment, (>52.6%, mean-2SE from healthy controls, NPC Group 1; <52.6%, NPC Group 2). The patients in Group 2 showed a significant decrease of CD3+CD8+ T-cells, CD3+CD4+ T-cells and CD3+CD45RO+ memory T cells, and increase of CD3-CD16+ NK cells compared to Group 1 patients and healthy controls (P<0.001). EBV-specific T cell responses, were weaker in this group of patients and their tumor cells expressed lower levels of the EBV encoded latent membrane protein (LMP)-1 and HLA class II protein compared with the patients of NPC Group 1 (P<0.05) . Conclusion: These findings demonstrate that NPC patients could be distinguished on the basis of their immune status which will affect the efficacy of EBV-CTL immunotherapy.
Collapse
Affiliation(s)
- Jiang Li
- State Key Laboratory of Oncology in South China, Sun Yat-sen University Cancer Center, Guangzhou, China
| | | | | | | | | | | |
Collapse
|
158
|
Comoli P, Zecca M, Maccario R. Immunotherapy against EBV-lymphoma in recipients of HSCT. Expert Rev Hematol 2011; 3:625-32. [PMID: 21083478 DOI: 10.1586/ehm.10.56] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
Epstein-Barr virus (EBV)-associated lymphoproliferations represent life-threatening complications of hematopoietic stem cell transplantation. In the last decade, immunological therapeutic strategies that allow us to selectively abrogate the origin of lymphoproliferation, namely B-cell compartment or EBV antigen-expressing tumor cells, have significantly reduced treatment-related toxicity while maintaining equal or superior efficacy. A further implementation is the possibility of preventing disease occurrence by delivering immunotherapy in the presymptomatic phase, on the basis of EBV-DNA blood levels. Despite the excellent results, T-cell therapy with EBV-specific cytotoxic T-lymphocytes has but a marginal role in the treatment of these forms. Promising implementations are underway, including logistic solutions to extend T-cell therapy beyond academic centers, delineation of strategies aimed at simplifying/shortening production and targeting immune evasion mechanisms exerted by tumor cells.
Collapse
Affiliation(s)
- Patrizia Comoli
- Fondazione IRCCS Policlinico S. Matteo, viale Golgi 19, 27100 Pavia, Italy
| | | | | |
Collapse
|
159
|
Secondino S, Zecca M, Licitra L, Gurrado A, Schiavetto I, Bossi P, Locati L, Schiavo R, Basso S, Baldanti F, Maccario R, Locatelli F, Siena S, Pedrazzoli P, Comoli P. T-cell therapy for EBV-associated nasopharyngeal carcinoma: preparative lymphodepleting chemotherapy does not improve clinical results. Ann Oncol 2011; 23:435-41. [PMID: 21586688 DOI: 10.1093/annonc/mdr134] [Citation(s) in RCA: 54] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
BACKGROUND We and others have demonstrated that adoptive cell therapy with Epstein-Barr virus (EBV)-specific autologous cytotoxic T lymphocytes (CTLs) may control disease progression in patients with EBV-associated nasopharyngeal carcinoma (NPC). With the aim of favoring in vivo T-cell expansion, we optimized our cell therapy approach by administering higher doses of EBV-specific CTLs, following lymphodepleting chemotherapy. PATIENTS AND METHODS Eleven patients with EBV-related NPC in whom conventional treatment failed have been enrolled. Patients received nonmyeloablative lymphodepleting chemotherapy consisting of cyclophosphamide and fludarabine. Two doses of autologous EBV-specific CTLs were subsequently infused, 2 weeks apart. Study end points were feasibility and clinical outcome. RESULTS All patients enrolled completed the treatment and were assessable for analysis. The median dose of CTLs per infusion was 3.7 × 10(8). Therapy was well tolerated, with no severe adverse events ascribable to either chemotherapy or cell therapy. Disease control (defined as either tumor regression or disease stabilization lasting >4 months) was obtained in 6 of 11 patients, in keeping with previously published results. CONCLUSIONS Our data confirm that EBV-specific CTL therapy is safe and associated with antitumor activity in patients with advanced NPC. The use of lymphodepleting chemotherapy before high-dose CTL infusion did not enhance the clinical benefit observed in our previous series.
Collapse
Affiliation(s)
- S Secondino
- Falck Medical Oncology, Niguarda Ca' Granda Hospital, Milano, Italy
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
160
|
Affiliation(s)
- Stephen Gottschalk
- Center for Cell and Gene Therapy, Texas Children's Cancer Center, Department of Pediatrics, Baylor College of Medicine, Texas Children's Hospital, The Methodist Hospital, Houston, Texas, USA
| |
Collapse
|
161
|
Quintarelli C, Dotti G, Hasan ST, De Angelis B, Hoyos V, Errichiello S, Mims M, Luciano L, Shafer J, Leen AM, Heslop HE, Rooney CM, Pane F, Brenner MK, Savoldo B. High-avidity cytotoxic T lymphocytes specific for a new PRAME-derived peptide can target leukemic and leukemic-precursor cells. Blood 2011; 117:3353-62. [PMID: 21278353 PMCID: PMC3069675 DOI: 10.1182/blood-2010-08-300376] [Citation(s) in RCA: 79] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2010] [Accepted: 01/01/2011] [Indexed: 11/20/2022] Open
Abstract
The cancer testis antigen (CTA) preferentially expressed antigen of melanoma (PRAME) is overexpressed by many hematologic malignancies, but is absent on normal tissues, including hematopoietic progenitor cells, and may therefore be an appropriate candidate for T cell-mediated immunotherapy. Because it is likely that an effective antitumor response will require high-avidity, PRAME-specific cytotoxic T lymphocytes (CTLs), we attempted to generate such CTLs using professional and artificial antigen-presenting cells loaded with a peptide library spanning the entire PRAME protein and consisting of 125 synthetic pentadecapeptides overlapping by 11 amino acids. We successfully generated polyclonal, PRAME-specific CTL lines and elicited high-avidity CTLs, with a high proportion of cells recognizing a previously uninvestigated HLA-A*02-restricted epitope, P435-9mer (NLTHVLYPV). These PRAME-CTLs could be generated both from normal donors and from subjects with PRAME(+) hematologic malignancies. The cytotoxic activity of our PRAME-specific CTLs was directed not only against leukemic blasts, but also against leukemic progenitor cells as assessed by colony-forming-inhibition assays, which have been implicated in leukemia relapse. These PRAME-directed CTLs did not affect normal hematopoietic progenitors, indicating that this approach may be of value for immunotherapy of PRAME(+) hematologic malignancies.
Collapse
MESH Headings
- Amino Acid Sequence
- Antigens, Neoplasm/chemistry
- Antigens, Neoplasm/genetics
- Antigens, Neoplasm/immunology
- Antigens, Neoplasm/metabolism
- Blood Donors
- Cell Line, Tumor
- Cytotoxicity, Immunologic/immunology
- Epitopes, T-Lymphocyte/chemistry
- Epitopes, T-Lymphocyte/immunology
- Epitopes, T-Lymphocyte/metabolism
- HLA-A Antigens/metabolism
- HLA-A2 Antigen
- Humans
- K562 Cells
- Leukemia/genetics
- Leukemia/immunology
- Leukemia/pathology
- Neoplastic Stem Cells/immunology
- Neoplastic Stem Cells/metabolism
- Peptide Fragments/chemistry
- Peptide Fragments/immunology
- T-Cell Antigen Receptor Specificity/immunology
- T-Cell Antigen Receptor Specificity/physiology
- T-Lymphocytes, Cytotoxic/immunology
- T-Lymphocytes, Cytotoxic/metabolism
Collapse
Affiliation(s)
- Concetta Quintarelli
- Center for Cell and Gene Therapy, Baylor College of Medicine, Houston, TX 77030, USA
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
162
|
Abstract
The outcome for patients with the most common primary brain tumor, glioblastoma multiforme (GBM), remains poor. Several immunotherapeutic approaches are actively being pursued including antibodies and cell-based therapies. While the blood-brain barrier protects brain tumor cells from therapeutic antibodies, immune cells have the ability to traverse the blood-brain barrier and migrate into GBM tumors to exert their therapeutic function. Results of Phase I clinical studies with vaccines to induce GBM-specific T cells are encouraging and Phase II clinical trials are in progress. Nonvaccine-based cell therapy for GBM has been actively explored over the last four decades. Here we will review past clinical experience with adoptive cell therapies for GBM and summarize current strategies on how to improve these approaches.
Collapse
MESH Headings
- Antigens, Neoplasm/chemistry
- Antigens, Neoplasm/immunology
- Clinical Trials as Topic
- Glioma/immunology
- Glioma/therapy
- Humans
- Immunotherapy, Adoptive/methods
- Immunotherapy, Adoptive/trends
- Killer Cells, Lymphokine-Activated/immunology
- Killer Cells, Natural/immunology
- Lymphocytes, Tumor-Infiltrating/immunology
- Receptors, Antigen, T-Cell, alpha-beta/genetics
- Receptors, Antigen, T-Cell, alpha-beta/immunology
- T-Lymphocytes, Cytotoxic/immunology
- Treatment Outcome
Collapse
Affiliation(s)
- K H Chow
- Texas Children's Cancer Center, Baylor College of Medicine, Houston, TX 77030, USA.
| | | |
Collapse
|
163
|
Merlo A, Turrini R, Dolcetti R, Zanovello P, Rosato A. Immunotherapy for EBV-associated malignancies. Int J Hematol 2011; 93:281-293. [PMID: 21336546 DOI: 10.1007/s12185-011-0782-2] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2010] [Revised: 01/10/2011] [Accepted: 02/01/2011] [Indexed: 02/03/2023]
Abstract
Since 1995 to date, more than 250 patients with EBV-related diseases received virus-specific CTL. Cell therapy proved to be safe and effective, and achieved some complete remissions also in patients who failed all previous standard treatments. The first clinical results with EBV-specific CTL were obtained for both prophylaxis and treatment of post-transplant lymphoproliferative disease arising in stem cell transplant or solid organ transplant recipients. Based on such encouraging results, the same approach was then extended to other EBV-related diseases, namely Hodgkin's lymphoma, nasopharyngeal carcinoma, and chronic active infection. Nowadays, the modification of the CTL generation protocols and the introduction of new specificities into EBV-specific CTL lines by chimeric antigen receptor transfer allow targeting other viral infections and also non-EBV related malignancies. Aim of this review is to summarize clinical results obtained thus far in adoptive cell therapy approaches with EBV-specific CTL. Moreover, by analyzing ongoing clinical trials, we also provide some insights on the potential future of a successful and paradigmatic history.
Collapse
Affiliation(s)
- Anna Merlo
- Department of Oncology and Surgical Sciences, University of Padova, Via Gattamelata, 64, 35128, Padua, Italy
| | - Riccardo Turrini
- Department of Oncology and Surgical Sciences, University of Padova, Via Gattamelata, 64, 35128, Padua, Italy
| | - Riccardo Dolcetti
- Cancer Bio-Immunotherapy Unit, Department of Medical Oncology, CRO, IRCCS, National Cancer Institute, Aviano, Italy
| | - Paola Zanovello
- Department of Oncology and Surgical Sciences, University of Padova, Via Gattamelata, 64, 35128, Padua, Italy.,Istituto Oncologico Veneto, IOV-IRCCS, Padua, Italy
| | - Antonio Rosato
- Department of Oncology and Surgical Sciences, University of Padova, Via Gattamelata, 64, 35128, Padua, Italy. .,Istituto Oncologico Veneto, IOV-IRCCS, Padua, Italy.
| |
Collapse
|
164
|
Paramita DK, Fatmawati C, Juwana H, van Schaijk FG, Fachiroh J, Haryana SM, Middeldorp JM. Humoral immune responses to Epstein-Barr virus encoded tumor associated proteins and their putative extracellular domains in nasopharyngeal carcinoma patients and regional controls. J Med Virol 2011; 83:665-78. [DOI: 10.1002/jmv.21960] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/07/2022]
|
165
|
Xu J, Wan XB, Huang XF, Chan KA, Hong MH, Wang LH, Long ZJ, Liu Q, Yan M, Lo YD, Zeng YX, Liu Q. Serologic Antienzyme Rate of Epstein-Barr Virus DNase-Specific Neutralizing Antibody Segregates TNM Classification in Nasopharyngeal Carcinoma. J Clin Oncol 2010; 28:5202-9. [DOI: 10.1200/jco.2009.25.6552] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
Purpose We investigate the value of pretreatment serologic antienzyme rate (AER) of Epstein-Barr virus (EBV) DNase-specific neutralizing antibody complementing TNM staging in prognostication of nasopharyngeal carcinoma (NPC). Patients and Methods Pretreatment serum samples from 1,303 patients with untreated NPC were collected and examined for AER. After a 10-year follow-up period, the prognoses of the patients, classified by their clinical stage with AER, were assessed by multivariate analysis. Of the 1,303 patients, 600 patients were randomly assigned to a training set to generate an AER cutoff point by receiver operating characteristic (ROC) curve analysis. AER levels were then analyzed with overall survival (OS), progression-free survival (PFS), local failure–free survival (LFFS), and distant metastasis–free survival (DMFS) in a testing set (703 patients). Another independent cohort of 464 patients was studied in a validating set. Results In the training set, the ROC analysis–generated AER cutoff point for OS was 58.0%, which was used as the cutoff point in the testing set. The subset of low AER levels predicted a significant survival advantage over the subset of high AER levels for OS, PFS, LFFS, and DMFS in the testing set. Moreover, two distinguished subgroups were segregated by an AER level of 58.0% within each clinical stage comparing prognostication of OS, PFS, LFFS, and DMFS. Importantly, AER level was revealed as the only significant independent prognostic factor for death, recurrence, and distant metastasis in the validating set. Conclusion Pretreatment serologic AER of EBV DNase-specific neutralizing antibody serves as an independent prognostic marker complementing TNM stage in NPC. Supplementing pretreatment AER with TNM staging leads to more accurate risk definition in patient subgroups.
Collapse
Affiliation(s)
- Jie Xu
- From the State Key Laboratory of Oncology in South China; Cancer Center; the Third Affiliated Hospital; Sun Yat-sen Institute of Hematology; Center for Clinical Trials and Institute of Drug Clinical Trials, Sun Yat-sen University, Guangzhou; and The Chinese University of Hong Kong, Hong Kong Special Administrative Region, China
| | - Xiang-Bo Wan
- From the State Key Laboratory of Oncology in South China; Cancer Center; the Third Affiliated Hospital; Sun Yat-sen Institute of Hematology; Center for Clinical Trials and Institute of Drug Clinical Trials, Sun Yat-sen University, Guangzhou; and The Chinese University of Hong Kong, Hong Kong Special Administrative Region, China
| | - Xue-Fei Huang
- From the State Key Laboratory of Oncology in South China; Cancer Center; the Third Affiliated Hospital; Sun Yat-sen Institute of Hematology; Center for Clinical Trials and Institute of Drug Clinical Trials, Sun Yat-sen University, Guangzhou; and The Chinese University of Hong Kong, Hong Kong Special Administrative Region, China
| | - K.C. Allen Chan
- From the State Key Laboratory of Oncology in South China; Cancer Center; the Third Affiliated Hospital; Sun Yat-sen Institute of Hematology; Center for Clinical Trials and Institute of Drug Clinical Trials, Sun Yat-sen University, Guangzhou; and The Chinese University of Hong Kong, Hong Kong Special Administrative Region, China
| | - Ming-Huang Hong
- From the State Key Laboratory of Oncology in South China; Cancer Center; the Third Affiliated Hospital; Sun Yat-sen Institute of Hematology; Center for Clinical Trials and Institute of Drug Clinical Trials, Sun Yat-sen University, Guangzhou; and The Chinese University of Hong Kong, Hong Kong Special Administrative Region, China
| | - Li-Hui Wang
- From the State Key Laboratory of Oncology in South China; Cancer Center; the Third Affiliated Hospital; Sun Yat-sen Institute of Hematology; Center for Clinical Trials and Institute of Drug Clinical Trials, Sun Yat-sen University, Guangzhou; and The Chinese University of Hong Kong, Hong Kong Special Administrative Region, China
| | - Zi-Jie Long
- From the State Key Laboratory of Oncology in South China; Cancer Center; the Third Affiliated Hospital; Sun Yat-sen Institute of Hematology; Center for Clinical Trials and Institute of Drug Clinical Trials, Sun Yat-sen University, Guangzhou; and The Chinese University of Hong Kong, Hong Kong Special Administrative Region, China
| | - Qing Liu
- From the State Key Laboratory of Oncology in South China; Cancer Center; the Third Affiliated Hospital; Sun Yat-sen Institute of Hematology; Center for Clinical Trials and Institute of Drug Clinical Trials, Sun Yat-sen University, Guangzhou; and The Chinese University of Hong Kong, Hong Kong Special Administrative Region, China
| | - Min Yan
- From the State Key Laboratory of Oncology in South China; Cancer Center; the Third Affiliated Hospital; Sun Yat-sen Institute of Hematology; Center for Clinical Trials and Institute of Drug Clinical Trials, Sun Yat-sen University, Guangzhou; and The Chinese University of Hong Kong, Hong Kong Special Administrative Region, China
| | - Y.M. Dennis Lo
- From the State Key Laboratory of Oncology in South China; Cancer Center; the Third Affiliated Hospital; Sun Yat-sen Institute of Hematology; Center for Clinical Trials and Institute of Drug Clinical Trials, Sun Yat-sen University, Guangzhou; and The Chinese University of Hong Kong, Hong Kong Special Administrative Region, China
| | - Yi-Xin Zeng
- From the State Key Laboratory of Oncology in South China; Cancer Center; the Third Affiliated Hospital; Sun Yat-sen Institute of Hematology; Center for Clinical Trials and Institute of Drug Clinical Trials, Sun Yat-sen University, Guangzhou; and The Chinese University of Hong Kong, Hong Kong Special Administrative Region, China
| | - Quentin Liu
- From the State Key Laboratory of Oncology in South China; Cancer Center; the Third Affiliated Hospital; Sun Yat-sen Institute of Hematology; Center for Clinical Trials and Institute of Drug Clinical Trials, Sun Yat-sen University, Guangzhou; and The Chinese University of Hong Kong, Hong Kong Special Administrative Region, China
| |
Collapse
|
166
|
Abstract
Current approaches for the treatment of tumours typically employ broad acting radiotherapeutic and chemotherapeutic approaches, which have led to high success rates but can be associated with unwanted side-effects. Cytotoxic T cell (CTL)-based immunotherapy offers an alternative approach that is designed to specifically target protein antigens expressed in malignant cells and is thus likely to limit any adverse side-effects. Defining tumour-specific antigens is therefore critical for the successful application of CTL-based therapy. Epstein-Barr virus (EBV)-associated malignancies offer an attractive target for CTL-based immunotherapy due to presence of virally encoded antigens in the malignant cells. Recent success in treating Epstein-Barr virus (EBV)-associated post-transplant lymphoproliferative disorder (PTLD) using cytotoxic T cell (CTL)-based immunotherapy has led to interest in the development of CTL-based immunotherapy to treat other EBV-associated malignancies in which antigen expression patterns are well defined but limited to a restricted number of proteins.
Collapse
|
167
|
Dotti G, Savoldo B, Brenner M. Fifteen years of gene therapy based on chimeric antigen receptors: "are we nearly there yet?". Hum Gene Ther 2010; 20:1229-39. [PMID: 19702437 DOI: 10.1089/hum.2009.142] [Citation(s) in RCA: 70] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022] Open
Abstract
"T-body" or chimeric antigen receptor (CAR) technology, which combines the specificity of an antibody with the homing, tissue penetration, and target cell destruction of T cells, was first described in 1993. After many years of unmet promise, significant improvements in gene transfer, including the development of efficient retroviral vectors for transduction of human T cells, and better understanding of immunological pathways and immune cell interactions, are allowing this technology to reach a critical phase of evaluation, in which we will learn whether the approach can truly meet expectations. In this review we summarize the concept of CAR-based immunotherapy, describe the steps accomplished, and outline the future progress we need to make if this approach is truly to improve cancer immunotherapy.
Collapse
Affiliation(s)
- Gianpietro Dotti
- Center for Cell and Gene Therapy, Baylor College of Medicine, Methodist Hospital, Houston, TX 77030, USA
| | | | | |
Collapse
|
168
|
Toh HC, Chia WK, Sun L, Thng CH, Soe Y, Phoon YP, Yap SP, Lim WT, Tai WM, Hee SW, Tan SH, Leong SS, Tan EH. Graft-vs-tumor effect in patients with advanced nasopharyngeal cancer treated with nonmyeloablative allogeneic PBSC transplantation. Bone Marrow Transplant 2010; 46:573-9. [PMID: 20661236 PMCID: PMC3072519 DOI: 10.1038/bmt.2010.161] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
While nonmyeloablative peripheral blood stem cell transplantation (NST) has shown efficacy against several solid tumors, it is untested in nasopharyngeal cancer (NPC). In a phase II clinical trial, 21 patients with pretreated metastatic NPC underwent NST with sibling PBSC allografts, using CY conditioning, thymic irradiation and in vivo T-cell depletion with thymoglobulin. Stable lymphohematopoietic chimerism was achieved in most patients and prophylactic CYA was tapered at a median of day +30. Seven patients (33%) showed partial response and three (14%) achieved stable disease. Four patients were alive at 2 years and three showed prolonged disease control of 344, 525 and 550 days. With a median follow-up of 209 (4-1147) days, the median PFS was 100 days (95% confidence interval (CI), 66-128 days), and median OS was 209 days (95% CI, 128-236 days). Patients with chronic GVHD had better survival-median OS 426 days (95% CI, 194-NE days) vs 143 days (95% CI, 114-226 days) (P=0.010). Thus, NST may induce meaningful clinical responses in patients with advanced NPC.
Collapse
Affiliation(s)
- H C Toh
- Department of Medical Oncology, National Cancer Centre, Singapore, Singapore.
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
169
|
Abstract
Early-stage nasopharyngeal carcinoma (T1-2a;N0;M0) represents a small proportion of nasopharyngeal tumors. Radiotherapy alone is the current treatment approach for this tumor and the emerging role of new radiotherapy techniques will hopefully further improve the treatment outcome for these patients. The vast majority of patients with nasopharyngeal carcinoma is diagnosed with locally advanced disease. Concomitant chemoradiotherapy is now acknowledged as being a standard treatment option, even though it induces a considerable incidence of acute mucosal and hematologic toxicity. The issue of adding adjuvant chemotherapy is somewhat more controversial. Similarly, the role of neoadjuvant chemotherapy before concomitant chemoradiotherapy is a matter of interest. In patients with recurrent/metastatic nasopharyngeal carcinoma the prognosis is generally grim, as platinum-based chemotherapy results in a 50-70% response rate and in a median survival time of 11 months. Several trials have been performed on this subset of patients with both cytotoxic and biologic agents, but the results have not been particularly encouraging thus far. Epstein-Barr virus is associated with the vast majority of nasopharyngeal carcinoma. Concentrations of plasma Epstein-Barr virus DNA have been associated with treatment outcome in the clinic. Immunotherapy is generally well tolerated and can sometimes elicit significant immune response, which possibly induces clinical benefit in some patients.
Collapse
|
170
|
Razak ARA, Siu LL, Liu FF, Ito E, O'Sullivan B, Chan K. Nasopharyngeal carcinoma: the next challenges. Eur J Cancer 2010; 46:1967-78. [PMID: 20451372 DOI: 10.1016/j.ejca.2010.04.004] [Citation(s) in RCA: 177] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2010] [Accepted: 04/06/2010] [Indexed: 12/12/2022]
Abstract
Nasopharyngeal carcinoma (NPC) differs from other head and neck cancers in its aetiology, epidemiology and potential therapeutic options. Despite cure for the majority of the patients, challenges still exist in the prevention of disease relapse, treatment of patients with refractory or metastatic NPC and the management of long-term toxicities. This article discusses the specific challenges in pushing the boundaries of NPC treatments further, with an emphasis on prognostic/predictive markers, molecularly targeted therapies, immunotherapies and the areas of interest with regard to long-term toxicities arising from therapeutic interventions.
Collapse
Affiliation(s)
- Albiruni R A Razak
- Division of Medical Oncology and Haematology, Princess Margaret Hospital, University of Toronto, Toronto, Ontario, Canada
| | | | | | | | | | | |
Collapse
|
171
|
Merlo A, Turrini R, Dolcetti R, Martorelli D, Muraro E, Comoli P, Rosato A. The interplay between Epstein-Barr virus and the immune system: a rationale for adoptive cell therapy of EBV-related disorders. Haematologica 2010; 95:1769-77. [PMID: 20421267 DOI: 10.3324/haematol.2010.023689] [Citation(s) in RCA: 77] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/15/2023] Open
Abstract
The Epstein-Barr virus has evolved a plethora of strategies to evade immune system recognition and to establish latent infection in memory B cells, where the virus resides lifelong without any consequence in the majority of individuals. However, some imbalances in the equilibrium between the inherent virus transforming properties and the host immune system can lead to the development of different tumors, such as lymphoproliferative disorders, Hodgkin's lymphoma, Burkitt's lymphoma, and nasopharyngeal carcinoma. The expression of viral antigens in malignant cells makes them suitable targets for immunotherapeutic approaches, which are mainly based on the ex vivo expansion of EBV-specific T cells. Indeed, the infusion of virus-specific cytotoxic T lymphocytes has proved not only to be safe and effective, but also capable of restoring or inducing a protective anti-virus immunity, which is lacking, albeit to a different extent, in every EBV-driven malignancy. The purpose of this review is to summarize the results of adoptive immunotherapy approaches for EBV-related malignancies, with particular emphasis on the immunological and virological aspects linked to the clinical responses obtained. Data collected confirm the clinical relevance of the use of EBV-specific cytotoxic T lymphocytes in the field of adoptive immunotherapy and suggest the increasing importance of this approach also against other tumors, concurrent with the increasing knowledge of the intimate and continuous interplay between the virus and the host immune system.
Collapse
Affiliation(s)
- Anna Merlo
- Department of Oncology and Surgical Sciences, University of Padova, Via Gattamelata 64, Padova, Italy
| | | | | | | | | | | | | |
Collapse
|
172
|
|
173
|
Merlo A, Turrini R, Bobisse S, Zamarchi R, Alaggio R, Dolcetti R, Mautner J, Zanovello P, Amadori A, Rosato A. Virus-specific cytotoxic CD4+ T cells for the treatment of EBV-related tumors. THE JOURNAL OF IMMUNOLOGY 2010; 184:5895-902. [PMID: 20385879 DOI: 10.4049/jimmunol.0902850] [Citation(s) in RCA: 44] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/04/2023]
Abstract
Although adoptive immunotherapy with CD8(+) CTL is providing clinically relevant results against EBV-driven malignancies, the effector role of CD4(+) T cells has been poorly investigated. We addressed this issue in a lymphoblastoid cell line-induced mouse model of posttransplant lymphoproliferative disease (PTLD) by comparing the therapeutic efficacy of EBV-specific CD4(+) and CD8(+) T cell lines upon adoptive transfer. CD4(+) T cells disclosed a long-lasting and stronger proliferative potential than CD8(+) T cells, had a similar activation and differentiation marker profile, efficiently killed their targets in a MHC class II-restricted manner, and displayed a lytic machinery comparable to that of cognate CD8(+) T cells. A detailed analysis of Ag specificity revealed that CD4(+) T cells potentially target EBV early lytic cycle proteins. Nonetheless, when assessed for the relative therapeutic impact after in vivo transfer, CD4(+) T cells showed a reduced activity compared with the CD8(+) CTL counterpart. This feature was apparently due to a strong and selective downmodulation of MHC class II expression on the tumor cells surface, a phenomenon that could be reverted by the demethylating agent 5-aza-2'-deoxycytidine, thus leading to restoration of lymphoblastoid cell line recognition and killing by CD4(+) T cells, as well as to a more pronounced therapeutic activity. Conversely, immunohistochemical analysis disclosed that HLA-II expression is fully retained in human PTLD samples. Our data indicate that EBV-specific cytotoxic CD4(+) T cells are therapeutic in mice bearing PTLD-like tumors, even in the absence of CD8(+) T cells. These findings pave the way to use cultures of pure CD4(+) T cells in immunotherapeutic approaches for EBV-related malignancies.
Collapse
Affiliation(s)
- Anna Merlo
- Department of Oncology and Surgical Sciences, University of Padova, Italy
| | | | | | | | | | | | | | | | | | | |
Collapse
|
174
|
Vera JF, Brenner LJ, Gerdemann U, Ngo MC, Sili U, Liu H, Wilson J, Dotti G, Heslop HE, Leen AM, Rooney CM. Accelerated production of antigen-specific T cells for preclinical and clinical applications using gas-permeable rapid expansion cultureware (G-Rex). J Immunother 2010; 33:305-15. [PMID: 20445351 PMCID: PMC2946348 DOI: 10.1097/cji.0b013e3181c0c3cb] [Citation(s) in RCA: 130] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
The clinical manufacture of antigen-specific cytotoxic T lymphocytes (CTLs) for adoptive immunotherapy is limited by the complexity and time required to produce large numbers with the desired function and specificity. The culture conditions required are rigorous, and in some cases only achieved in 2-cm wells in which cell growth is limited by gas exchange, nutrients, and waste accumulation. Bioreactors developed to overcome these issues tend to be complex, expensive, and not always conducive to CTL growth. We observed that antigen-specific CTLs undergo 7 to 10 divisions poststimulation. However, the expected CTL numbers were achieved only in the first week of culture. By recreating the culture conditions present during this first week-low frequency of antigen-specific T cells and high frequency of feeder cells-we were able to increase CTL expansion to expected levels that could be sustained for several weeks without affecting phenotype or function. However, the number of 24-well plates needed was excessive and cultures required frequent media changes, increasing complexity and manufacturing costs. Therefore, we evaluated novel gas-permeable culture devices (G-Rex) with a silicone membrane at the base allowing gas exchange to occur uninhibited by the depth of the medium above. This system effectively supports the expansion of CTL and actually increases output by up to 20-fold while decreasing the required technician time. Importantly, this amplified cell expansion is not because of more cell divisions but because of reduced cell death. This bioprocess optimization increased T-cell output while decreasing the complexity and cost of CTL manufacture, making cell therapy more accessible.
Collapse
Affiliation(s)
- Juan F Vera
- Center for Cell and Gene Therapy, Baylor College of Medicine, Houston, TX 77030, USA.
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
175
|
Emergence of BCR-ABL–specific cytotoxic T cells in the bone marrow of patients with Ph+ acute lymphoblastic leukemia during long-term imatinib mesylate treatment. Blood 2010; 115:1512-8. [PMID: 20007806 DOI: 10.1182/blood-2009-06-230391] [Citation(s) in RCA: 40] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Abstract
Imatinib mesylate has been demonstrated to allow the emergence of T cells directed against chronic myeloid leukemia cells. A total of 10 Philadelphia chromosome–positive acute lymphoblastic leukemia patients receiving high-dose imatinib mesylate maintenance underwent long-term immunological monitoring (range, 2-65 months) of p190BCR-ABL–specific T cells in the bone marrow and peripheral blood. p190BCR-ABL–specific T lymphocytes were detected in all patients, more frequently in bone marrow than in peripheral blood samples (67% vs 25%, P < .01) and resulted significantly associated with lower minimal residual disease values (P < .001), whereas absent at leukemia relapse. Specific T cells were mainly effector memory CD8+ and CD4+ T cells, producing interferon-γ, tumor necrosis factor-α, and interleukin-2 (median percentage of positive cells: 3.34, 3.04, and 3.58, respectively). Cytotoxic subsets able to lyse BCR-ABL–positive leukemia blasts also were detectable. Whether these autologous p190BCR-ABL–specific T cells may be detectable under other tyrosine-kinase inhibitors, expanded ex vivo, and exploited for immunotherapy remains to be addressed.
Collapse
|
176
|
T cell receptor gene therapy: strategies for optimizing transgenic TCR pairing. Trends Mol Med 2010; 16:77-87. [DOI: 10.1016/j.molmed.2009.12.004] [Citation(s) in RCA: 76] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2009] [Revised: 12/21/2009] [Accepted: 12/21/2009] [Indexed: 11/19/2022]
|
177
|
Novel approach to the formulation of an Epstein-Barr virus antigen-based nasopharyngeal carcinoma vaccine. J Virol 2010; 84:407-17. [PMID: 19846527 DOI: 10.1128/jvi.01303-09] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Epstein-Barr virus (EBV) is associated with several malignant diseases including nasopharyngeal carcinoma (NPC), a common neoplasm throughout southeast Asia. Radiotherapy and chemotherapy can achieve remission, but a reemergence of disease is not uncommon. Therefore, there is a need for specific therapies that target the tumor through the recognition of EBV antigens. In NPC, latent membrane protein 1 (LMP1) and LMP2 offer the best opportunity for specific targeting since they are typically expressed and T-cell determinants in each of these proteins have been defined. We have attempted to maximize the opportunity of incorporating every possible CD4 and CD8 determinant in a single formulation. We have achieved this by generating a scrambled protein incorporating random overlapping peptide sets from EBNA1, LMP1, and LMP2, which was then inserted into a replication-deficient strain of adenovirus (adenovirus scrambled antigen vaccine [Ad-SAVINE]). This report describes the construction of this Ad-SAVINE construct, its utility in generating LMP1 and LMP2 responses in healthy individuals as well as NPC patients, and its capacity to define new epitopes. This formulation could have a role in NPC immunotherapy for all ethnic groups since it has the potential to activate all possible CD4 and CD8 responses within EBNA1 and LMPs.
Collapse
|
178
|
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
|
179
|
Immunotherapy of head and neck cancer: current and future considerations. JOURNAL OF ONCOLOGY 2009; 2009:346345. [PMID: 19680453 PMCID: PMC2723756 DOI: 10.1155/2009/346345] [Citation(s) in RCA: 41] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 02/17/2009] [Accepted: 06/15/2009] [Indexed: 01/09/2023]
Abstract
Patients with head and neck squamous cell carcinoma (HNSCC) are at considerable risk for death, with 5-year relative survival rates of approximately 60%. The profound multifaceted deficiencies in cell-mediated immunity that persist in most patients after treatment may be related to the high rates of treatment failure and second primary malignancies. Radiotherapy and chemoradiotherapy commonly have severe acute and long-term side effects on immune responses. The development of immunotherapies reflects growing awareness that certain immune system deficiencies specific to HNSCC and some other cancers may contribute to the poor long-term outcomes. Systemic cell-mediated immunotherapy is intended to activate the entire immune system and mount a systemic and/or locoregional antitumor response. The delivery of cytokines, either by single cytokines, for example, interleukin-2, interleukin-12, interferon-γ, interferon-α, or by a biologic mix of multiple cytokines, such as IRX-2, may result in tumor rejection and durable immune responses. Targeted immunotherapy makes use of monoclonal antibodies or vaccines. All immunotherapies for HNSCC except cetuximab remain investigational, but a number of agents whose efficacy and tolerability are promising have entered phase 2 or phase 3 development.
Collapse
|
180
|
Dotti G, Tian M, Savoldo B, Najjar A, Cooper LJ, Jackson J, Smith A, Mawlawi O, Uthamanthil R, Borne A, Brammer D, Paolillo V, Alauddin M, Gonzalez C, Steiner D, Decker WK, Marini F, Kornblau S, Bollard CM, Shpall EJ, Gelovani JG. Repetitive Noninvasive Monitoring of HSV1-tk-Expressing T Cells Intravenously Infused into Nonhuman Primates Using Positron Emission Tomography and Computed Tomography with
18
F-FEAU. Mol Imaging 2009. [DOI: 10.2310/7290.2009.00022] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Affiliation(s)
- Gianpietro Dotti
- From the Departments of Experimental Diagnostic Imaging, Nuclear Medicine, Imaging Physics, Veterinary Medicine and Surgery, Stem Cell Transplantation and Cellular Therapy, and Pediatrics, The University of Texas M.D. Anderson Cancer Center, and the Center for Cell and Gene Therapy, Baylor College of Medicine, Houston, TX
| | - Mei Tian
- From the Departments of Experimental Diagnostic Imaging, Nuclear Medicine, Imaging Physics, Veterinary Medicine and Surgery, Stem Cell Transplantation and Cellular Therapy, and Pediatrics, The University of Texas M.D. Anderson Cancer Center, and the Center for Cell and Gene Therapy, Baylor College of Medicine, Houston, TX
| | - Barbara Savoldo
- From the Departments of Experimental Diagnostic Imaging, Nuclear Medicine, Imaging Physics, Veterinary Medicine and Surgery, Stem Cell Transplantation and Cellular Therapy, and Pediatrics, The University of Texas M.D. Anderson Cancer Center, and the Center for Cell and Gene Therapy, Baylor College of Medicine, Houston, TX
| | - Amer Najjar
- From the Departments of Experimental Diagnostic Imaging, Nuclear Medicine, Imaging Physics, Veterinary Medicine and Surgery, Stem Cell Transplantation and Cellular Therapy, and Pediatrics, The University of Texas M.D. Anderson Cancer Center, and the Center for Cell and Gene Therapy, Baylor College of Medicine, Houston, TX
| | - Laurence J.N. Cooper
- From the Departments of Experimental Diagnostic Imaging, Nuclear Medicine, Imaging Physics, Veterinary Medicine and Surgery, Stem Cell Transplantation and Cellular Therapy, and Pediatrics, The University of Texas M.D. Anderson Cancer Center, and the Center for Cell and Gene Therapy, Baylor College of Medicine, Houston, TX
| | - James Jackson
- From the Departments of Experimental Diagnostic Imaging, Nuclear Medicine, Imaging Physics, Veterinary Medicine and Surgery, Stem Cell Transplantation and Cellular Therapy, and Pediatrics, The University of Texas M.D. Anderson Cancer Center, and the Center for Cell and Gene Therapy, Baylor College of Medicine, Houston, TX
| | - Amanda Smith
- From the Departments of Experimental Diagnostic Imaging, Nuclear Medicine, Imaging Physics, Veterinary Medicine and Surgery, Stem Cell Transplantation and Cellular Therapy, and Pediatrics, The University of Texas M.D. Anderson Cancer Center, and the Center for Cell and Gene Therapy, Baylor College of Medicine, Houston, TX
| | - Osama Mawlawi
- From the Departments of Experimental Diagnostic Imaging, Nuclear Medicine, Imaging Physics, Veterinary Medicine and Surgery, Stem Cell Transplantation and Cellular Therapy, and Pediatrics, The University of Texas M.D. Anderson Cancer Center, and the Center for Cell and Gene Therapy, Baylor College of Medicine, Houston, TX
| | - Rajesh Uthamanthil
- From the Departments of Experimental Diagnostic Imaging, Nuclear Medicine, Imaging Physics, Veterinary Medicine and Surgery, Stem Cell Transplantation and Cellular Therapy, and Pediatrics, The University of Texas M.D. Anderson Cancer Center, and the Center for Cell and Gene Therapy, Baylor College of Medicine, Houston, TX
| | - Agatha Borne
- From the Departments of Experimental Diagnostic Imaging, Nuclear Medicine, Imaging Physics, Veterinary Medicine and Surgery, Stem Cell Transplantation and Cellular Therapy, and Pediatrics, The University of Texas M.D. Anderson Cancer Center, and the Center for Cell and Gene Therapy, Baylor College of Medicine, Houston, TX
| | - David Brammer
- From the Departments of Experimental Diagnostic Imaging, Nuclear Medicine, Imaging Physics, Veterinary Medicine and Surgery, Stem Cell Transplantation and Cellular Therapy, and Pediatrics, The University of Texas M.D. Anderson Cancer Center, and the Center for Cell and Gene Therapy, Baylor College of Medicine, Houston, TX
| | - Vincenzo Paolillo
- From the Departments of Experimental Diagnostic Imaging, Nuclear Medicine, Imaging Physics, Veterinary Medicine and Surgery, Stem Cell Transplantation and Cellular Therapy, and Pediatrics, The University of Texas M.D. Anderson Cancer Center, and the Center for Cell and Gene Therapy, Baylor College of Medicine, Houston, TX
| | - Mian Alauddin
- From the Departments of Experimental Diagnostic Imaging, Nuclear Medicine, Imaging Physics, Veterinary Medicine and Surgery, Stem Cell Transplantation and Cellular Therapy, and Pediatrics, The University of Texas M.D. Anderson Cancer Center, and the Center for Cell and Gene Therapy, Baylor College of Medicine, Houston, TX
| | - Carlos Gonzalez
- From the Departments of Experimental Diagnostic Imaging, Nuclear Medicine, Imaging Physics, Veterinary Medicine and Surgery, Stem Cell Transplantation and Cellular Therapy, and Pediatrics, The University of Texas M.D. Anderson Cancer Center, and the Center for Cell and Gene Therapy, Baylor College of Medicine, Houston, TX
| | - David Steiner
- From the Departments of Experimental Diagnostic Imaging, Nuclear Medicine, Imaging Physics, Veterinary Medicine and Surgery, Stem Cell Transplantation and Cellular Therapy, and Pediatrics, The University of Texas M.D. Anderson Cancer Center, and the Center for Cell and Gene Therapy, Baylor College of Medicine, Houston, TX
| | - William K. Decker
- From the Departments of Experimental Diagnostic Imaging, Nuclear Medicine, Imaging Physics, Veterinary Medicine and Surgery, Stem Cell Transplantation and Cellular Therapy, and Pediatrics, The University of Texas M.D. Anderson Cancer Center, and the Center for Cell and Gene Therapy, Baylor College of Medicine, Houston, TX
| | - Frank Marini
- From the Departments of Experimental Diagnostic Imaging, Nuclear Medicine, Imaging Physics, Veterinary Medicine and Surgery, Stem Cell Transplantation and Cellular Therapy, and Pediatrics, The University of Texas M.D. Anderson Cancer Center, and the Center for Cell and Gene Therapy, Baylor College of Medicine, Houston, TX
| | - Steven Kornblau
- From the Departments of Experimental Diagnostic Imaging, Nuclear Medicine, Imaging Physics, Veterinary Medicine and Surgery, Stem Cell Transplantation and Cellular Therapy, and Pediatrics, The University of Texas M.D. Anderson Cancer Center, and the Center for Cell and Gene Therapy, Baylor College of Medicine, Houston, TX
| | - Catherine M. Bollard
- From the Departments of Experimental Diagnostic Imaging, Nuclear Medicine, Imaging Physics, Veterinary Medicine and Surgery, Stem Cell Transplantation and Cellular Therapy, and Pediatrics, The University of Texas M.D. Anderson Cancer Center, and the Center for Cell and Gene Therapy, Baylor College of Medicine, Houston, TX
| | - Elizabeth J. Shpall
- From the Departments of Experimental Diagnostic Imaging, Nuclear Medicine, Imaging Physics, Veterinary Medicine and Surgery, Stem Cell Transplantation and Cellular Therapy, and Pediatrics, The University of Texas M.D. Anderson Cancer Center, and the Center for Cell and Gene Therapy, Baylor College of Medicine, Houston, TX
| | - Juri G. Gelovani
- From the Departments of Experimental Diagnostic Imaging, Nuclear Medicine, Imaging Physics, Veterinary Medicine and Surgery, Stem Cell Transplantation and Cellular Therapy, and Pediatrics, The University of Texas M.D. Anderson Cancer Center, and the Center for Cell and Gene Therapy, Baylor College of Medicine, Houston, TX
| |
Collapse
|
181
|
Lutzky VP, Davis JE, Crooks P, Corban M, Smith MC, Elliott M, Morrison L, Cross S, Tscharke D, Panizza B, Coman W, Bharadwaj M, Moss DJ. Optimization of LMP-specific CTL expansion for potential adoptive immunotherapy in NPC patients. Immunol Cell Biol 2009; 87:481-8. [PMID: 19468283 DOI: 10.1038/icb.2009.25] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Nasopharyngeal carcinoma (NPC) is Epstein-Barr virus (EBV) positive in all undifferentiated cases, expressing the latency II phenotype of latent membrane proteins (LMPs) 1 and 2, in addition to EBV nuclear antigen (EBNA) 1. Several studies have attempted to treat NPC with EBV-specific cytotoxic T lymphocyte (CTL) with a partial response. To improve this therapy, there is a need to expand CTL targeted to the latency II antigens of EBV, rather than the immunodominant EBV nuclear antigens 3-6 peptides typically expanded by lymphoblastoid cells. In order to maximize the expansion of LMP-specific CTL in vitro for use in adoptive immunotherapy of nasopharyngeal carcinoma patients, we used lymphoblastoid cell lines coated with synthetic peptides corresponding to CTL determinants from the LMP proteins. We investigated several issues pertaining to the expansion of an immunologically weak CTL response, including peptide and interleukin-2 concentration, and screening assays for selecting the optimal peptide for use in expansion of LMP-specific CTL. Although screening of ex vivo peripheral blood mononuclear cells did not prove to be useful in the selection of an LMP peptide for use in CTL cultures, the peptide and interleukin-2 concentrations were critical for the maximum expansion of CTL. Therefore, it is imperative that stimulation protocols are optimized for the expansion of LMP-specific CTL.
Collapse
Affiliation(s)
- Viviana P Lutzky
- EBV Biology Laboratory, Division of Immunology, Australian Centre for Vaccine Development, The Queensland Institute of Medical Research, The Royal Brisbane Hospital, 300 Herston Road, Herston, Brisbane, QLD 4006, Australia.
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
182
|
Vera JF, Hoyos V, Savoldo B, Quintarelli C, Giordano Attianese GMP, Leen AM, Liu H, Foster AE, Heslop HE, Rooney CM, Brenner MK, Dotti G. Genetic manipulation of tumor-specific cytotoxic T lymphocytes to restore responsiveness to IL-7. Mol Ther 2009; 17:880-8. [PMID: 19259067 PMCID: PMC2835146 DOI: 10.1038/mt.2009.34] [Citation(s) in RCA: 79] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2008] [Accepted: 02/03/2009] [Indexed: 12/17/2022] Open
Abstract
Adoptive transfer of antigen-specific cytotoxic T lymphocytes (CTLs) can induce objective clinical responses in patients with malignant diseases. The option of providing a proliferative and survival advantage to adoptively transferred CTLs remains a challenge to improve their efficacy. Host lymphodepletion and administration of recombinant interleukin-2 (IL-2) are currently used to improve CTL survival and expansion after adoptive transfer, but these approaches are frequently associated with significant side effects and may increase proliferation of T regulatory cells. IL-7 is a crucial homeostatic cytokine that has been safely administered as a recombinant protein. However, while IL-7 induces robust expansion of naive and memory T lymphocytes, the lack of expression of the IL-7 receptor alpha chain (IL-7Ralpha) by CTLs precludes their response to this cytokine. We found that CTLs can be genetically modified to re-express IL-7Ralpha, and that this manipulation restores the response of these cells to IL-7 without apparent modification of their antigen specificity or dependency, and without changing their response to other common gamma (gammac) chain cytokines. This approach may allow selective expansion of CTLs without the unwanted effects associated with IL-2.
Collapse
Affiliation(s)
- Juan F Vera
- Center for Cell and Gene Therapy, Baylor College of Medicine, The Methodist Hospital, Houston, Texas 77030, USA
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
183
|
Louis CU, Straathof K, Bollard CM, Gerken C, Huls MH, Gresik MV, Wu MF, Weiss HL, Gee AP, Brenner MK, Rooney CM, Heslop HE, Gottschalk S. Enhancing the in vivo expansion of adoptively transferred EBV-specific CTL with lymphodepleting CD45 monoclonal antibodies in NPC patients. Blood 2009; 113:2442-50. [PMID: 18971421 PMCID: PMC2656271 DOI: 10.1182/blood-2008-05-157222] [Citation(s) in RCA: 90] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2008] [Accepted: 10/15/2008] [Indexed: 12/16/2022] Open
Abstract
Treatment of Epstein-Barr virus (EBV)-positive nasopharyngeal carcinoma (NPC) with EBV-specific cytotoxic T cells (EBV-specific CTL) has been promising, producing clinical responses. However, infused EBV-specific CTL did not expand in vivo, likely limiting their antitumor activity. Lymphodepleting patients with chemotherapy before T-cell transfer enhances in vivo T-cell expansion, but results in nonspecific destruction of the resident immune system and can have significant toxicity. To evaluate if monoclonal antibodies (mAbs) can produce a more selective lymphodepletion, we conducted a clinical study in which NPC patients received a pair of lymphodepleting mAbs targeted to the CD45 antigen (CD45 mAbs) before EBV-specific CTL infusion. Eight patients with recurrent NPC received CD45 mAbs followed by escalating doses of autologous EBV-specific CTL. Infusion of CD45 mAbs resulted in transient lymphopenia in all patients and an increase in interleukin-15 (IL-15) levels in 6 out 8 patients. All patients had an increase in their peripheral blood frequency of EBV-specific T cells after CTL infusion. Three patients with a persistent increase had clinical benefits including 1 complete response (> 24 months) and 2 with stable disease (for 12 and 15 months). Lymphodepleting mAbs prior CTL transfer may represent an alternative to chemotherapy to enhance expansion of infused CTL. This study is registered at (http://www.clinialtrials.gov) as NCT00608257.
Collapse
Affiliation(s)
- Chrystal U Louis
- Center for Cell and Gene Therapy, Baylor College of Medicine, Houston, TX 77030, USA
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
184
|
Lu H, Peng L, Yuan X, Hao Y, Lu Z, Chen J, Cheng J, Deng S, Gu J, Pang Q, Qin J. Concurrent chemoradiotherapy in locally advanced nasopharyngeal carcinoma: a treatment paradigm also applicable to patients in Southeast Asia. Cancer Treat Rev 2009; 35:345-53. [PMID: 19211192 DOI: 10.1016/j.ctrv.2009.01.002] [Citation(s) in RCA: 43] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2008] [Revised: 01/05/2009] [Accepted: 01/07/2009] [Indexed: 11/17/2022]
Abstract
The majority of nasopharyngeal carcinoma (NPC) patients present at locally advanced stage. The poor prognosis has led to increasing interests in exploring the use of chemotherapy (CT). Intergroup-0099 trial was the first randomized trial comparing concurrent chemoradiotherapy (CCRT) with radiotherapy (RT) alone. Its outcome established the treatment standard in the United States as standard of care for locally advanced NPC. However, criticism has been arisen, particularly about its reproducibility and applicability in Southeast Asia where NPC is an endemic disease. Subsequently, new evidence has been provided by a large number of publications from various centers. In this article, through comprehensively analyzing recent meta-analyses and randomized controlled trials performed in Asian centers, we conclude that CCRT as a treatment paradigm is also applicable to patients in Southeast Asia and should be standard of practice in locally advanced disease. However, the CT regimen varied markedly among those trials, and the optimal regimen and scheduling remains to be determined. Moreover, a number of patients experienced toxicities and the treatment compliance was generally poor. With the emergence of new RT techniques such as intensity-modulated radiotherapy (IMRT) and image-guided radiotherapy (IGRT), the role of concurrent CT with these new techniques needs to be tested. New chemotherapeutics have been investigated in the recurrent or metastatic disease. However, their effectiveness in previously untreated NPC is unclear. Previous efforts have been made for immunotherapy and targeted therapy in palliative setting. Their role in newly diagnosed NPC should be evaluated, particularly when they are combined with CT or RT.
Collapse
Affiliation(s)
- Heming Lu
- Department of Radiation Oncology, People's Hospital of Guangxi Zhuang Autonomous Region, Nanning City, PR China.
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
185
|
Abstract
This chapter describes the major gene therapeutic approaches for viral infections. The vast majority of published approaches target severe chronic viral infections such as hepatitis B or C and HIV infection. Two basic gene therapy strategies are introduced here. The first involves the expression of a protein or an RNA that inhibits viral replication by targeting crucial steps of the viral life cycle or by interfering with a cellular factor required for virus replication. The major limitation of this approach is that primary levels of gene modification have generally not been sufficient to reduce the availability of target cells permissive for virus replication to a level that significantly decreases overall viral load. Thus, investigators have banked on the expectation that gene-protected cells have a sufficient selective advantage to accumulate and gain prevalence over time, a prediction that so far could not be confirmed in clinical trials. In vivo levels of gene modification can be improved, however, by introducing an additional selectable marker. In addition, a secreted antiviral gene product that exerts a bystander effect could significantly reduce overall virus replication despite relatively low levels of gene modification. In addition to these direct antiviral approaches, several strategies have been developed that employ or aim to enhance host immune responses. The innate immune response has been enhanced, for example, by the in vivo expression of interferons. Alternatively, T cells can be grafted with recombinant receptors to boost adaptive virus-specific immunity. These approaches are especially promising for chronic virus infection, where natural immune responses are evidently not sufficient to effectively control virus replication.
Collapse
|
186
|
Merlo A, Turrini R, Dolcetti R, Zanovello P, Amadori A, Rosato A. Adoptive cell therapy against EBV-related malignancies: a survey of clinical results. Expert Opin Biol Ther 2008; 8:1265-94. [PMID: 18694349 DOI: 10.1517/14712598.8.9.1265] [Citation(s) in RCA: 31] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
Abstract
BACKGROUND Epstein-Barr Virus (EBV) infection is associated with a heterogeneous group of tumors, including lymphoproliferative disorders, Hodgkin's disease, nasopharyngeal carcinoma and Burkitt's lymphoma. As such neoplastic disorders express viral antigens, they can be treated by adoptive immunotherapy strategies relying mostly on in vitro generation and expansion of virus-specific cytotoxic T lymphocytes (CTL), which can be administered to patients for both prophylaxis and treatment. OBJECTIVE We reviewed results obtained in all clinical trials reported thus far employing anti-EBV adoptive immunotherapy for different virus-related malignancies. METHODS 'PTLD after HSCT', 'PTLD after SOT', 'NPC', 'HD', 'SCAEBV' and 'extranodal NK/T cell lymphoma', in combination with 'Adoptive immunotherapy' and 'Adoptive transfer', were used as search keys for papers in PubMed. CONCLUSIONS Although the heterogeneity of different studies precludes their collection for a meta-analysis, it can be inferred that adoptive therapy with EBV-specific CTL is safe, well tolerated and particularly effective in the case of most immunogenic tumors, like post-transplant lymphoproliferative disease.
Collapse
Affiliation(s)
- Anna Merlo
- University of Padova, Department of Oncology and Surgical Sciences, Via Gattamelata 64, I-35128 Padova, Italy
| | | | | | | | | | | |
Collapse
|
187
|
Effect of frequently used chemotherapeutic drugs on cytotoxic activity of human cytotoxic T-lymphocytes. J Immunother 2008; 31:283-93. [PMID: 18317359 DOI: 10.1097/cji.0b013e3181628b76] [Citation(s) in RCA: 31] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023]
Abstract
Tumors are considered to be possible targets of immunotherapy using stimulated and expanded cytotoxic T-lymphocytes (CTL). It is important to consider the drug-induced effects when chemotherapeutic regimens and CTL-mediated immunotherapy is planned to be used in parallel. In this study, we characterized the effect of 29 frequently used chemotherapeutic agents on the cytotoxic activity of autologous and allogeneic CTLs. We found that treatment of CTLs with the following drugs: docetaxel, vincristine, chlorambucil, mitomycin C, oxaliplatin, doxorubicin, and bleomycin effectively inhibited CTL-mediated killing, without affecting their viability. On the other hand, the following drugs enhanced or permitted efficient CTL-mediated killing in vitro at concentrations comparable with the maximally achieved therapeutic concentration in vivo in humans: daunorubicin, prednisolone, vinorelbine, cisplatin, methotrexate, hydroxyurea, cytarabine, cyclophosphamide, topotecan, epirubicin, fluorouracil, carboplatin, asparaginase, 6-mercaptopurine, and bortezomib. Our results could potentially be used in the future to design new CTL-based adjuvant immunotherapy protocols.
Collapse
|
188
|
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: 156] [Impact Index Per Article: 9.2] [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
|
189
|
Brenner MK. Developing T-cell therapies for cancer in an academic setting. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2008; 610:88-99. [PMID: 18593017 DOI: 10.1007/978-0-387-73898-7_7] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/16/2023]
Abstract
T-cell immunotherapies have great potential because of their exquisite targeting ability and high efficacy, but are more complex to implement since they do not readily fit the pharmaceutical model. Nonetheless, recent improvements in gene-transfer technology allow T cells directed to tumor-associated antigens to effectively eradicate even bulky malignancies. T lymphocytes also have the potential to target tumor stem cells, provided the target antigens are appropriately identified and expressed on the components used to generate the effector T-cells. Numerous problems remain before these approaches can be widely implemented, but as the success rate increases, willingness to commit the necessary resources will correspondingly increase.
Collapse
Affiliation(s)
- Malcolm K Brenner
- Center for Cell and Gene Therapy, Baylor College of Medicine, The Methodist Hospital, Texas Children's Hospital, TX, USA.
| |
Collapse
|
190
|
Martorelli D, Houali K, Caggiari L, Vaccher E, Barzan L, Franchin G, Gloghini A, Pavan A, Da Ponte A, Tedeschi RM, De Re V, Carbone A, Ooka T, De Paoli P, Dolcetti R. Spontaneous T cell responses to Epstein-Barr virus-encoded BARF1 protein and derived peptides in patients with nasopharyngeal carcinoma: bases for improved immunotherapy. Int J Cancer 2008; 123:1100-7. [PMID: 18546263 DOI: 10.1002/ijc.23621] [Citation(s) in RCA: 28] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Abstract
Immunotherapy approaches targeting Epstein-Barr virus (EBV)-encoded antigens induce objective clinical responses only in a fraction of patients with undifferentiated nasopharyngeal carcinoma (UNPC). In the present study, we have characterized the immunogenicity of the EBV-encoded BARF1 oncogene with the aim to assess whether this protein could constitute a new target antigen for immunotherapy in this setting. Spontaneous CD4+ and CD8+ T cell responses specific for the recombinant p29 BARF1 protein were detected by IFNgamma-ELISPOT in both EBV-seropositive donors and UNPC patients, but not in EBV-seronegative individuals. Using immunoinformatic prediction tools, we have selected 5 different candidate BARF1 T cell epitopes presented by HLA-A*0201. Although only one of these peptides was able to bind HLA-A2 with low affinity in the T2 stabilization assay, all 5 BARF1 nonamers readily elicited specific CD8+ T cell responses in EBV-seropositive HLA-A*0201+ donors and UNPC patients. Notably, the magnitude of CD8+ T cell responses to the whole BARF1 protein and derived A*0201 peptides was significantly higher in UNPC patients than in healthy donors. Moreover, cytotoxic T lymphocytes specific for the p2-10, p23-31, or p49-57 BARF1 peptides were easily obtained from HLA-A*0201+ donors. These cultures were not only able to lyse autologous targets loaded with the antigenic peptide, but also recognized tumor cells endogenously expressing BARF1 in an antigen-specific and HLA-A2-restricted manner. These findings, indicate that BARF1 is a particularly attractive antigen with immunogenic properties in most UNPC patients and provide valuable information to develop new strategies to improve the efficacy of EBV-targeting immunotherapy of UNPC patients.
Collapse
Affiliation(s)
- Debora Martorelli
- Cancer Bioimmunotherapy Unit, Centro di Riferimento Oncologico, IRCCS-National Cancer Institute, Aviano, PN, Italy
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
191
|
Wee J. 4th FY Khoo Memorial Lecture 2008: Nasopharyngeal Cancer Workgroup – The Past, The Present and The Future. ANNALS OF THE ACADEMY OF MEDICINE, SINGAPORE 2008. [DOI: 10.47102/annals-acadmedsg.v37n7p606] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/09/2023]
|
192
|
Ma BBY, Hui EP, Chan ATC. Systemic approach to improving treatment outcome in nasopharyngeal carcinoma: current and future directions. Cancer Sci 2008; 99:1311-8. [PMID: 18498420 PMCID: PMC11158701 DOI: 10.1111/j.1349-7006.2008.00836.x] [Citation(s) in RCA: 93] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2008] [Revised: 03/22/2008] [Accepted: 03/27/2008] [Indexed: 12/13/2022] Open
Abstract
Systemic therapy is an integral part of the management of non-keratinizing nasopharyngeal carcinoma (NPC). The purposes of this review are to provide the latest results and future directions of clinical and translational research for this disease, and to illustrate how some of these new therapies have improved the treatment outcome for patients with NPC. Particular attention will be paid to the clinical application of chemotherapy in the adjunctive treatment of locoregionally advanced NPC, novel targeted drugs, Epstein-Barr virus-targeted vaccine therapies, and the use of plasma Epstein-Barr virus DNA as a biomarker for selecting patients for adjunctive therapies.
Collapse
Affiliation(s)
- Brigette B Y Ma
- Department of Clinical Oncology at the Sir Y. K. Pao Center for Cancer, Prince of Wales Hospital, Ngan Shing Street, Shatin, New Territories, Hong Kong SAR, China
| | | | | |
Collapse
|
193
|
Lin X, Gudgeon NH, Hui EP, Jia H, Qun X, Taylor GS, Barnardo MCNM, Lin CK, Rickinson AB, Chan ATC. CD4 and CD8 T cell responses to tumour-associated Epstein-Barr virus antigens in nasopharyngeal carcinoma patients. Cancer Immunol Immunother 2008; 57:963-75. [PMID: 18094968 PMCID: PMC11031094 DOI: 10.1007/s00262-007-0427-8] [Citation(s) in RCA: 36] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2007] [Accepted: 11/10/2007] [Indexed: 01/14/2023]
Abstract
Nasopharyngeal carcinoma (NPC), an Epstein-Barr virus (EBV)-associated tumour common in Southern Chinese populations, is a potentially important target for T cell-based immunotherapy. The tumour cells are HLA class I- and II-positive and express a limited subset of EBV latent proteins, namely the nuclear antigen EBNA1 and the latent membrane proteins LMP2 and (in some cases) LMP1. To ask whether the tumour develops in the presence of a potentially protective host response or in its absence, we set out to determine the prevailing levels of CD4+ and CD8+ T cell memory to these proteins in NPC patients at tumour diagnosis. We first screened healthy Chinese donors against Chinese strain EBNA1, LMP1 and LMP2 sequences in Elispot assays of interferon-gamma release and identified the immunodominant CD4+ and CD8+ epitope peptides presented by common Chinese HLA alleles. Then, comparing 60 patients with >70 healthy controls on peptide epitope mini-panels, we found that T cell memory to CD4 epitopes in all three proteins was unimpaired in the blood of patients at diagnosis. In most cases NPC patients also showed detectable responses to CD8 epitopes relevant to their HLA type, the one consistent exception being the absence in patients of a B*4001-restricted response to LMP2. We infer that NPC arises in patients whose prevailing levels of T cell memory to tumour-associated EBV proteins is largely intact; the therapeutic goal must therefore be to re-direct the existing memory repertoire more effectively against antigen-expressing tumour cells.
Collapse
Affiliation(s)
- Xiaorong Lin
- Sir Y. K. Pao Centre for Cancer, Hong Kong Cancer Institute, The Chinese University of Hong Kong, Hong Kong, China
| | - Nancy H. Gudgeon
- Cancer Research UK Institute for Cancer Studies, University of Birmingham, Vincent Drive, Birmingham, B15 2TT UK
| | - Edwin P. Hui
- Sir Y. K. Pao Centre for Cancer, Hong Kong Cancer Institute, The Chinese University of Hong Kong, Hong Kong, China
- Department of Clinical Oncology, The Chinese University of Hong Kong, Hong Kong, China
| | - Hui Jia
- Cancer Research UK Institute for Cancer Studies, University of Birmingham, Vincent Drive, Birmingham, B15 2TT UK
| | - Xue Qun
- Sir Y. K. Pao Centre for Cancer, Hong Kong Cancer Institute, The Chinese University of Hong Kong, Hong Kong, China
| | - Graham S. Taylor
- Cancer Research UK Institute for Cancer Studies, University of Birmingham, Vincent Drive, Birmingham, B15 2TT UK
| | - Martin C. N. M. Barnardo
- Department of Transplant Immunology, Oxford Transplant Centre, University of Oxford, Churchill Hospital, Oxford, UK
| | - C. Kit Lin
- Hong Kong Red Cross Blood Transfusion Service, Kowloon, Hong Kong, China
| | - Alan B. Rickinson
- Cancer Research UK Institute for Cancer Studies, University of Birmingham, Vincent Drive, Birmingham, B15 2TT UK
| | - Anthony T. C. Chan
- Sir Y. K. Pao Centre for Cancer, Hong Kong Cancer Institute, The Chinese University of Hong Kong, Hong Kong, China
- Department of Clinical Oncology, The Chinese University of Hong Kong, Hong Kong, China
| |
Collapse
|
194
|
Conrad H, Gebhard K, Krönig H, Neudorfer J, Busch DH, Peschel C, Bernhard H. CTLs Directed against HER2 Specifically Cross-React with HER3 and HER4. THE JOURNAL OF IMMUNOLOGY 2008; 180:8135-45. [DOI: 10.4049/jimmunol.180.12.8135] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
|
195
|
Kasamon YL, Ambinder RF. Immunotherapies for Hodgkin's lymphoma. Crit Rev Oncol Hematol 2008; 66:135-44. [PMID: 18023356 PMCID: PMC5792053 DOI: 10.1016/j.critrevonc.2007.10.001] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2007] [Revised: 09/19/2007] [Accepted: 10/03/2007] [Indexed: 11/17/2022] Open
Abstract
Multiple immune evasion strategies characterize the pathobiology of Hodgkin's lymphoma. These must be considered when developing and testing immunotherapeutic approaches for this disease. The clinical experience with adoptive immunotherapy of Epstein-Barr virus positive tumors, and with monoclonal antibodies directed against CD30, CD20, and other antigens, is herein reviewed.
Collapse
Affiliation(s)
- Yvette L Kasamon
- Division of Hematologic Malignancies, Johns Hopkins Medical Institutions, Baltimore, MD, USA.
| | | |
Collapse
|
196
|
Rosenberg SA, Restifo NP, Yang JC, Morgan RA, Dudley ME. Adoptive cell transfer: a clinical path to effective cancer immunotherapy. Nat Rev Cancer 2008; 8:299-308. [PMID: 18354418 PMCID: PMC2553205 DOI: 10.1038/nrc2355] [Citation(s) in RCA: 1208] [Impact Index Per Article: 71.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Adoptive cell therapy (ACT) using autologous tumour-infiltrating lymphocytes has emerged as the most effective treatment for patients with metastatic melanoma and can mediate objective cancer regression in approximately 50% of patients. The use of donor lymphocytes for ACT is an effective treatment for immunosuppressed patients who develop post-transplant lymphomas. The ability to genetically engineer human lymphocytes and use them to mediate cancer regression in patients, which has recently been demonstrated, has opened possibilities for the extension of ACT immunotherapy to patients with a wide variety of cancer types and is a promising new approach to cancer treatment.
Collapse
Affiliation(s)
- Steven A Rosenberg
- Surgery Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, 10 Center Drive, Bethesda, Maryland 20892, USA.
| | | | | | | | | |
Collapse
|
197
|
Craddock J, Heslop HE. Adoptive cellular therapy with T cells specific for EBV-derived tumor antigens. UPDATE ON CANCER THERAPEUTICS 2008; 3:33-41. [PMID: 19255606 PMCID: PMC2390887 DOI: 10.1016/j.uct.2008.01.001] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 01/28/2023]
Affiliation(s)
- John Craddock
- Center for Cell and Gene Therapy, Baylor College of Medicine, The Methodist Hospital and Texas Children's Hospital, Houston, TX
| | | |
Collapse
|
198
|
Curti A, Tosi P, Comoli P, Terragna C, Ferri E, Cellini C, Massaia M, D'Addio A, Giudice V, Di Bello C, Cavo M, Conte R, Gugliotta G, Baccarani M, Lemoli RM. Phase I/II clinical trial of sequential subcutaneous and intravenous delivery of dendritic cell vaccination for refractory multiple myeloma using patient-specific tumour idiotype protein or idiotype (VDJ)-derived class I-restricted peptides. Br J Haematol 2008; 139:415-24. [PMID: 17910631 DOI: 10.1111/j.1365-2141.2007.06832.x] [Citation(s) in RCA: 46] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022]
Abstract
Fifteen multiple myeloma (MM) patients who had failed maintenance therapy after tandem autologous stem cell transplantation underwent anti-idiotype (Id) vaccination with dendritic cells (DCs). CD14(+)-derived DCs were loaded with the autologous Id as whole protein (=6) or Id-derived class I-restricted peptides (=9) and keyhole limpet hemocyanin (KLH). Vaccination consisted of three subcutaneous (sc) and two intravenous injections of increasing DC doses at 2 weeks interval. DC therapy was well tolerated. Most patients developed both humoral and T-cell responses to KLH, suggesting immunocompetence. Eight of 15 patients developed an Id-specific T-cell proliferative response, 8/15 increased interferon-gamma-secreting T cells and 4/15 showed an Id-positive delayed-type hypersensitivity test. Anti-Id cytotoxic T-lymphocyte precursors increased after DC vaccination in 2/2 evaluable patients. A more robust T-cell response was observed after sc DC injections and increased Id-specific T-cell proliferation was found up to 1 year after vaccination. VDJ-derived peptides were as effective as the whole protein in stimulating T-cell responses. Clinically, 7/15 patients have stable disease after a median follow-up of 26 months, one patient achieved durable partial remission after 40 months, and seven patients progressed. In conclusion, sc injections of cryopreserved Id-pulsed DCs were safe and, in contrast with intravenous administrations, induced anti-MM T-cell responses.
Collapse
Affiliation(s)
- Antonio Curti
- Institute of Haematology and Medical Oncology 'L. & A. Seràgnoli', University of Bologna, Bologna, Italy
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
199
|
Abstract
The human hepatitis B virus is a small, enveloped and non-cytopathic virus, with a very narrow host range and a strong liver tropism causing acute and chronic liver disease. Although a well-tolerated vaccine is available, more than 350 million people are chronically infected worldwide. Available therapies for chronic hepatitis B only rarely eliminate the virus. Although new antivirals are being developed, long-term treatment is required, which may be limited by the selection of resistant viruses. Therefore, immunotherapies are investigated as an approach to eliminate persistently infected cells. Besides therapeutic vaccination, adoptive T-cell therapy is an interesting option, which is discussed in this review.
Collapse
Affiliation(s)
- F Bohne
- Centre for Molecular Medicine, Institute for Medical Microbiology, Immunology and Hygiene, University of Cologne, Cologne, Germany
| | | |
Collapse
|
200
|
Quintarelli C, Vera JF, Savoldo B, Giordano Attianese GMP, Pule M, Foster AE, Heslop HE, Rooney CM, Brenner MK, Dotti G. Co-expression of cytokine and suicide genes to enhance the activity and safety of tumor-specific cytotoxic T lymphocytes. Blood 2007; 110:2793-802. [PMID: 17638856 PMCID: PMC2018664 DOI: 10.1182/blood-2007-02-072843] [Citation(s) in RCA: 133] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2007] [Accepted: 07/12/2007] [Indexed: 01/22/2023] Open
Abstract
The antitumor effect of adoptively transferred tumor-specific cytotoxic T lymphocytes (CTLs) is impaired by the limited capacity of these cells to expand within the tumor microenvironment. Administration of interleukin 2 (IL-2) has been used to overcome this limitation, but the systemic toxicity and the expansion of unwanted cells, including regulatory T cells, limit the clinical value of this strategy. To discover whether transgenic expression of lymphokines by the CTLs themselves might overcome these limitations, we evaluated the effects of transgenic expression of IL-2 and IL-15 in our model of Epstein Barr Virus-specific CTLs (EBV-CTLs). We found that transgenic expression of IL-2 or IL-15 increased the expansion of EBV-CTLs both in vitro and in vivo in a severe combined immunodeficiency disease (SCID) mouse model and enhanced antitumor activity. Although the proliferation of these cytokine genes transduced CTLs remained strictly antigen dependent, clinical application of this approach likely requires the inclusion of a suicide gene to deal with the potential development of T-cell mutants with autonomous growth. We found that the incorporation of an inducible caspase-9 suicide gene allowed efficient elimination of transgenic CTLs after exposure to a chemical inducer of dimerization, thereby increasing the safety and feasibility of the approach.
Collapse
Affiliation(s)
- Concetta Quintarelli
- Center for Cell and Gene Therapy, Baylor College of Medicine, Houston, TX 77030, USA
| | | | | | | | | | | | | | | | | | | |
Collapse
|