251
|
McLean M, Wallace HL, Sharma A, Hill HC, Sabel MS, Egilmez NK. A BALB/c murine lung alveolar carcinoma used to establish a surgical spontaneous metastasis model. Clin Exp Metastasis 2004; 21:363-9. [PMID: 15554393 DOI: 10.1023/b:clin.0000046176.33867.c5] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023]
Abstract
Line-1, a weakly immunogenic lung tumor cell line derived from the BALB/c mouse, metastasizes spontaneously to the lungs of mice following subcutaneous administration. The parameters that influence metastasis as well as the progression of metastatic lung disease following surgical resection of primary subcutaneous tumors were characterized. Histological analysis of the lungs obtained from mice bearing different size subcutaneous tumors demonstrated that >90% of the mice developed micrometastatic disease in the lungs when the tumor exceeded 650 mm3 in size. Surgical resection of subcutaneous tumors resulted in the cure of primary disease in 95% of the mice. Macroscopic tumor nodules were grossly visible in the lungs of 75% of the mice 5 weeks after surgery. Serum amyloid A level correlated with primary tumor burden and was diagnostic for the presence of metastatic disease. The efficiency of metastasis, post-surgical primary tumor recurrence and long-term survival were significantly different between BALB/c mice obtained from different suppliers. The Line-1-BALB/c surgical metastasis model provides a clinically relevant tool for the evaluation of anti-cancer therapies, especially those that are designed to target long-term suppression of minimal residual disease following surgical intervention.
Collapse
Affiliation(s)
- Michael McLean
- Department of Microbiology and Immunology, State University of New York at Buffalo, Buffalo, New York, USA
| | | | | | | | | | | |
Collapse
|
252
|
Mocellin S, Semenzato G, Mandruzzato S, Rossi CR. Part II: Vaccines for haematological malignant disorders. Lancet Oncol 2004; 5:727-37. [DOI: 10.1016/s1470-2045(04)01649-3] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/25/2022]
|
253
|
Lundin KU, Screpanti V, Omholt H, Hofgaard PO, Yagita H, Grandien A, Bogen B. CD4+ T cells kill Id+ B-lymphoma cells: FasLigand-Fas interaction is dominant in vitro but is redundant in vivo. Cancer Immunol Immunother 2004; 53:1135-45. [PMID: 15696611 PMCID: PMC11032948 DOI: 10.1007/s00262-004-0538-4] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2004] [Accepted: 03/19/2004] [Indexed: 01/30/2023]
Abstract
B-lymphoma cells express a highly tumor-specific antigen, monoclonal Ig, which is a promising target for immunotherapy. Previous work has demonstrated that B-lymphoma cells spontaneously process their endogenous monoclonal Ig and present variable (V) region peptides (Id-peptides) on their MHC class II molecules to CD4+ T cells. Id-specific CD4+ T cells protect mice against B-lymphoma cells in the absence of antiidiotypic antibodies. The molecular mechanism by which Id-specific CD4+ T cells kill B-lymphoma cells is hitherto unknown. We here demonstrate in an Id-specific T-cell receptor (TCR)-transgenic mouse model that Id-specific CD4+ T cells induce apoptosis of Fas+ B-lymphoma cells in vitro by FasLigand (FasL)-Fas interaction. Moreover, the rare B lymphomas that had escaped rejection in TCR-transgenic mice had down-regulated their sensitivity to Fas-mediated apoptosis. Although these results suggest that FasL-Fas interaction is important, Id-specific CD4+ T cells could eliminate Id+ B-lymphoma cells in vivo by other mechanisms, since three independent ways of blocking FasL-Fas-mediated killing failed to abrogate tumor protection in TCR-transgenic mice. These results suggest that there are several redundant pathways by which Id-specific CD4+ T cells eliminate Id+ B-lymphoma cells in vivo, of which FasL-Fas interaction is only one.
Collapse
Affiliation(s)
- Katrin U Lundin
- Institute of Immunology, University of Oslo, Rikshospitalet, N-0027 Oslo, Norway.
| | | | | | | | | | | | | |
Collapse
|
254
|
Weng WK, Czerwinski D, Timmerman J, Hsu FJ, Levy R. Clinical Outcome of Lymphoma Patients After Idiotype Vaccination Is Correlated With Humoral Immune Response and Immunoglobulin G Fc Receptor Genotype. J Clin Oncol 2004; 22:4717-24. [PMID: 15483014 DOI: 10.1200/jco.2004.06.003] [Citation(s) in RCA: 161] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
Purpose The unique immunoglobulin idiotype (Id) expressed by each B-cell lymphoma is a target for immunotherapy. Vaccination with Id induces humoral and/or cellular anti-Id immune responses. However, the clinical impact of these anti-Id immune responses is unknown. We and others have previously reported that immunoglobulin G Fc receptor (FcγR) polymorphisms predict the clinical response of lymphoma patients to passive anti-CD20 antibody infusions. In this study, we tested whether anti-Id immune responses or FcγR polymorphisms associate with clinical outcome of patients who received Id vaccination. Patients and Methods We analyzed 136 patients with follicular lymphoma who had received Id vaccination. The anti-Id immune responses were measured and FcγRIIIa and FcγRIIa polymorphisms were determined and correlated with clinical outcome for these patients. Results Patients who mounted humoral immune responses had a longer progression-free survival (PFS) than those who did not (8.21 v 3.38 years; P = .018). Patients with FcγRIIIa 158 valine/valine (V/V) genotype also had a longer PFS than those with valine/phenylalanine (V/F) or phenylalanine/phenylalanine (F/F) genotypes (V/V, 8.21 v V/F, 3.38 years; P = .004; v F/F, 4.47 years; P = .035). Multivariate analysis using the Cox proportional hazards model showed that V/V genotype and humoral immune responses were independent positive predictors for PFS. Conclusion This study is the first to identify the predictive value of FcγR polymorphism on clinical outcome in patients who received active immunotherapy with tumor antigen vaccines. Our results imply that the antibodies induced against a tumor antigen are beneficial and that FcγR-bearing cells mediate an antitumor effect by killing antibody-coated tumor cells.
Collapse
MESH Headings
- Antigens, CD/genetics
- Antigens, CD/metabolism
- Cohort Studies
- Female
- Genetic Markers
- Humans
- Immunoglobulin Idiotypes/therapeutic use
- Immunotherapy, Active/methods
- Lymphoma, B-Cell/genetics
- Lymphoma, B-Cell/immunology
- Lymphoma, B-Cell/mortality
- Lymphoma, B-Cell/therapy
- Lymphoma, Follicular/genetics
- Lymphoma, Follicular/immunology
- Lymphoma, Follicular/mortality
- Lymphoma, Follicular/therapy
- Lymphoma, Non-Hodgkin/genetics
- Lymphoma, Non-Hodgkin/immunology
- Lymphoma, Non-Hodgkin/mortality
- Lymphoma, Non-Hodgkin/therapy
- Male
- Multivariate Analysis
- Polymorphism, Genetic
- Predictive Value of Tests
- Prognosis
- Proportional Hazards Models
- Prospective Studies
- Receptors, IgG/genetics
- Receptors, IgG/metabolism
- Risk Assessment
- Sensitivity and Specificity
- Survival Analysis
- Treatment Outcome
- Vaccination/methods
Collapse
Affiliation(s)
- Wen-Kai Weng
- Division of Oncology, Department of Internal Medicine, Stanford University School of Medicine, Stanford, CA 94305-5306, USA
| | | | | | | | | |
Collapse
|
255
|
Dave SS, Wright G, Tan B, Rosenwald A, Gascoyne RD, Chan WC, Fisher RI, Braziel RM, Rimsza LM, Grogan TM, Miller TP, LeBlanc M, Greiner TC, Weisenburger DD, Lynch JC, Vose J, Armitage JO, Smeland EB, Kvaloy S, Holte H, Delabie J, Connors JM, Lansdorp PM, Ouyang Q, Lister TA, Davies AJ, Norton AJ, Muller-Hermelink HK, Ott G, Campo E, Montserrat E, Wilson WH, Jaffe ES, Simon R, Yang L, Powell J, Zhao H, Goldschmidt N, Chiorazzi M, Staudt LM. Prediction of survival in follicular lymphoma based on molecular features of tumor-infiltrating immune cells. N Engl J Med 2004; 351:2159-69. [PMID: 15548776 DOI: 10.1056/nejmoa041869] [Citation(s) in RCA: 998] [Impact Index Per Article: 49.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/21/2023]
Abstract
BACKGROUND Patients with follicular lymphoma may survive for periods of less than 1 year to more than 20 years after diagnosis. We used gene-expression profiles of tumor-biopsy specimens obtained at diagnosis to develop a molecular predictor of the length of survival. METHODS Gene-expression profiling was performed on 191 biopsy specimens obtained from patients with untreated follicular lymphoma. Supervised methods were used to discover expression patterns associated with the length of survival in a training set of 95 specimens. A molecular predictor of survival was constructed from these genes and validated in an independent test set of 96 specimens. RESULTS Individual genes that predicted the length of survival were grouped into gene-expression signatures on the basis of their expression in the training set, and two such signatures were used to construct a survival predictor. The two signatures allowed patients with specimens in the test set to be divided into four quartiles with widely disparate median lengths of survival (13.6, 11.1, 10.8, and 3.9 years), independently of clinical prognostic variables. Flow cytometry showed that these signatures reflected gene expression by nonmalignant tumor-infiltrating immune cells. CONCLUSIONS The length of survival among patients with follicular lymphoma correlates with the molecular features of nonmalignant immune cells present in the tumor at diagnosis.
Collapse
|
256
|
Wooldridge JE, Weiner GJ. CpG DNA and cancer immunotherapy: orchestrating the antitumor immune response. Curr Opin Oncol 2004; 15:440-5. [PMID: 14624226 DOI: 10.1097/00001622-200311000-00006] [Citation(s) in RCA: 39] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
PURPOSE OF REVIEW Cancer treatment is entering an era of targeted approaches. One such approach is use of the immune system to recognize and eliminate malignant cells. Synthetic CpG oligonucleotides (CpG DNA) are a relatively new class of agents that have the ability to stimulate a potent, orchestrated tumor-specific immune response. This review provides an overview of the immunologic effects of CpG DNA and summarizes the results of preclinical investigations that have led to ongoing development of CpG DNA as a component of clinical cancer immunotherapy. RECENT FINDINGS New studies demonstrate that at least three classes of CpG DNA sequences exist, each with different physical characteristics and biologic effects. Preliminary studies in several animal models of cancer suggest that CpG DNA have the ability to induce tumor regression by activating innate immunity, enhancing antibody dependent cellular cytotoxicity, and serving as potent vaccine adjuvants that elicit a specific, protective immune response. SUMMARY Animal models suggest that CpG DNA may have many uses in cancer immunotherapy. Early clinical trials suggest that CpG DNA can be administered safely to humans, and studies are ongoing to understand how these agents may play a role in cancer immunotherapy.
Collapse
Affiliation(s)
- James E Wooldridge
- Department of Internal Medicine, Holden Comprehensive Cancer Center, University of Iowa, Iowa City, 52242, USA.
| | | |
Collapse
|
257
|
Abstract
Twelve years after the first formal demonstration that it is possible to vaccinate a cancer patient against an antigen derived from his/her own tumor, idiotype vaccines are now well into Phase III clinical trials for the treatment of follicular lymphoma. Meanwhile, their potential has also begun to be explored in other non-Hodgkin's lymphoma settings, such as that of mantle cell lymphoma. Another well known field of potential application for idiotype vaccines is that of multiple myeloma. However, the currently available results, even with the advent of dendritic cells, seem to be less promising than those obtained in lymphoma, to such an extent that idiotype vaccines are currently tested in multiple myeloma patients in the context of more aggressive therapeutic strategies.
Collapse
MESH Headings
- Antibodies, Anti-Idiotypic/immunology
- Cancer Vaccines/immunology
- Dendritic Cells, Follicular/immunology
- Humans
- Immunoglobulin Idiotypes/immunology
- Lymphoma, B-Cell/immunology
- Lymphoma, B-Cell/prevention & control
- Lymphoma, Follicular/immunology
- Lymphoma, Follicular/prevention & control
- Lymphoma, Mantle-Cell/immunology
- Lymphoma, Mantle-Cell/prevention & control
- Lymphoma, Non-Hodgkin/immunology
- Lymphoma, Non-Hodgkin/prevention & control
- Multiple Myeloma/immunology
- Multiple Myeloma/prevention & control
- Vaccines, DNA/immunology
Collapse
Affiliation(s)
- Maurizio Bendandi
- Department of Hematology and Cell Therapy Area, University Clinic of Navarre - Center for Applied Medical Research, Avda. Pio XII, 36 31008 Pamplona (Navarre), Spain.
| |
Collapse
|
258
|
von Bergwelt-Baildon M, Maecker B, Schultze J, Gribben JG. CD40 activation: potential for specific immunotherapy in B-CLL. Ann Oncol 2004; 15:853-7. [PMID: 15151939 DOI: 10.1093/annonc/mdh213] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023] Open
Abstract
Despite encouraging scientific and therapeutic advances, chronic lymphocytic leukemia (CLL) principally remains an incurable disease. Allogeneic transplantation represents the only curative approach, but is marked by high mortality. Novel and less toxic treatment modalities are needed. Immunotherapeutic approaches have clearly demonstrated potential effectiveness in CLL and other B-cell malignancies. To successfully direct immunity against CLL, highly immunogenic tumor cells or tumor-antigen-loaded antigen-presenting cells are necessary. The CD40-CD40L interaction has been shown to significantly increase antigen presentation in normal and malignant B-cells. Here we discuss biology and potential therapeutic applications of the CD40-system in CLL.
Collapse
Affiliation(s)
- M von Bergwelt-Baildon
- Department of Medical Oncology, Disease Center for Hematologic Neoplasia, Dana-Farber Cancer Institute and Harvard Medical School, Boston, MA 02115, USA
| | | | | | | |
Collapse
|
259
|
The APC's of tumor vaccine therapy. Blood 2004. [DOI: 10.1182/blood-2004-07-2644] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
|
260
|
Grube M, Rezvani K, Wiestner A, Fujiwara H, Sconocchia G, Melenhorst JJ, Hensel N, Marti GE, Kwak LW, Wilson W, Barrett JA. Autoreactive, cytotoxic T lymphocytes specific for peptides derived from normal B-cell differentiation antigens in healthy individuals and patients with B-cell malignancies. Clin Cancer Res 2004; 10:1047-56. [PMID: 14871984 DOI: 10.1158/1078-0432.ccr-03-0075] [Citation(s) in RCA: 17] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
PURPOSE To investigate potential immunotherapeutic strategies in B lymphocytic malignancies we looked for CTLs recognizing CD19 and CD20 epitopes. EXPERIMENTAL DESIGN Three CD19 and CD20 peptides binding to HLA-A*0201 were identified and used to detect peptide specific CTLs by a quantitative real-time PCR to measure IFN-gamma mRNA expression in 23 healthy individuals and 28 patients (18 chronic lymphocytic leukemia (CLL), 7 follicular lymphoma, 2 acute lymphocytic leukemia, and 1 large cell lymphoma). Peptide-specific CTLs were expanded in culture with CD40-activated B cells to test lytic activity in three patients. RESULTS In healthy individuals, CD8+ T-cell responses were detected in one to CD19(74-82), in three to CD20(127-135), and three to CD20(188-196). Seven of 27 patients (6 with CLL) had CD8+ T cells recognizing CD19(74-82). Seven patients responded to CD20(127-135) and three to CD20(188-196). All were CLL patients. CD19(74-82)-specific CTLs from three patients were expanded over 4 weeks. These cells were HLA-A*0201 specific and lytic for peptide-loaded antigen-presenting cells but not to malignant or unpulsed B cells. CONCLUSIONS CTLs that recognize CD19 and CD20 epitopes exist in healthy individuals and may be increased in CLL patients. They are of low avidity and require high doses of peptide for activation. Strategies to increase T-cell avidity would be necessary for T-cell immunotherapeutic approaches using the peptides studied.
Collapse
MESH Headings
- Adult
- Aged
- Aged, 80 and over
- Antigens/chemistry
- Antigens, CD19/biosynthesis
- Antigens, CD19/metabolism
- Antigens, CD20/biosynthesis
- Antigens, CD20/metabolism
- Antigens, Differentiation, B-Lymphocyte/immunology
- Antigens, Differentiation, B-Lymphocyte/metabolism
- B-Lymphocytes/immunology
- B-Lymphocytes/metabolism
- CD40 Antigens/metabolism
- CD8-Positive T-Lymphocytes/metabolism
- Cell Line, Tumor
- DNA, Complementary/metabolism
- Female
- Flow Cytometry
- HLA-A Antigens/metabolism
- HLA-A2 Antigen
- Humans
- Interferon-gamma/metabolism
- Lymphoma, B-Cell/immunology
- Lymphoma, B-Cell/metabolism
- Male
- Middle Aged
- Peptides/chemistry
- Polymerase Chain Reaction
- Protein Binding
- RNA/chemistry
- RNA, Messenger/metabolism
- Reverse Transcriptase Polymerase Chain Reaction
- T-Lymphocytes, Cytotoxic/metabolism
Collapse
Affiliation(s)
- Matthias Grube
- Hematology Branch, National Heart-Lung-Blood Institute/NIH, 9000 Rockville Pike, Building 10, Bethesda, MD 20892, USA
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
261
|
Spicer J, Quatan N, Plunkett T, Pandha H. Cellular immunotherapy for cancer: current concepts and clinical perspectives. Clin Oncol (R Coll Radiol) 2004; 16:395-404. [PMID: 15487131 DOI: 10.1016/j.clon.2004.03.014] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/26/2022]
Affiliation(s)
- J Spicer
- Department of Urology, St George's Hospital Medical School, London, UK
| | | | | | | |
Collapse
|
262
|
Dermime S, Gilham DE, Shaw DM, Davidson EJ, Meziane EK, Armstrong A, Hawkins RE, Stern PL. Vaccine and antibody-directed T cell tumour immunotherapy. Biochim Biophys Acta Rev Cancer 2004; 1704:11-35. [PMID: 15238242 DOI: 10.1016/j.bbcan.2004.03.002] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2003] [Revised: 01/22/2004] [Accepted: 03/31/2004] [Indexed: 10/26/2022]
Abstract
Clearer evidence for immune surveillance in malignancy and the identification of many new tumour-associated antigens (TAAs) have driven novel vaccine and antibody-targeted responses for therapy in cancer. The exploitation of active immunisation may be particularly favourable for TAA where tolerance is incomplete but passive immunisation may offer an additional strategy where the immune repertoire is affected by either tolerance or immune suppression. This review will consider how to utilise both active and passive types of therapy delivered by T cells in the context of the failure of tumour-specific immunity by presenting cancer patients. This article will outline the progress, problems and prospects of several different vaccine and antibody-targeted approaches for immunotherapy of cancer where proof of principle pre-clinical studies have been or will soon be translated into the clinic. Two examples of vaccination-based therapies where both T cell- and antibody-mediated anti-tumour responses are likely to be relevant and two examples of oncofoetal antigen-specific antibody-directed T cell therapies are described in the following sections: (1) therapeutic vaccination against human papillomavirus (HPV) antigens in cervical neoplasia; (2) B cell lymphoma vaccines including against immunoglobulin idiotype; (3) oncofoetal antigens as tumour targets for redirecting T cells with antibody strategies.
Collapse
Affiliation(s)
- Said Dermime
- Immunology, Cancer Research UK Groups, Paterson Institute for Cancer Research and University of Manchester, Christie Hospital NHS Trust, Manchester M20 4BX, UK
| | | | | | | | | | | | | | | |
Collapse
|
263
|
Suárez E, Yáñez R, Barrios Y, Díaz-Espada F. Human monoclonal antibodies produced in transgenic BABκ,λ mice recognising idiotypic immunoglobulins of human lymphoma cells. Mol Immunol 2004; 41:519-26. [PMID: 15183930 DOI: 10.1016/j.molimm.2004.03.035] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2004] [Indexed: 10/26/2022]
Abstract
Clonal idiotypic immunoglobulins of follicular lymphomas can be isolated by somatic fusion procedures. Idiotypic IgMs (Id-IgM) were isolated from two patients and used to immunise a strain of mice, deficient in mouse antibody production and engineered with yeast artificial chromosomes (YAC) containing fragments of the human immunoglobulin (Ig) micro/delta heavy chain and kappa/lambda light chain loci. Sequence analysis showed that hybridomas prepared from spleen cells of immunised mice expressed exclusively one of the six VH genes (VH1-2) present in the YAC transgene with different D/J rearrangements, and secrete fully human monoclonal antibodies (mAb) that recognised the tumour-specific IgM proteins. Further studies of the reactivity of the monoclonal anti-human Id-IgM antibodies revealed that they are specific for the individual protein of each patient and probably react with idiotypic determinants. In one case studied, the antibody recognised specifically the lymphoma cell expressing the corresponding idiotypic IgM and lysed those cells in the presence of complement. This is the first example of a human monoclonal antibody with such characteristics and may be of further use in the therapy of patients with B cell malignancies.
Collapse
Affiliation(s)
- Eduardo Suárez
- Department of Immunology, Hospital Clínica Puerta de Hierro, San Martín de Porres 4, Madrid 28035, Spain
| | | | | | | |
Collapse
|
264
|
Baskar S, Kobrin CB, Kwak LW. Autologous lymphoma vaccines induce human T cell responses against multiple, unique epitopes. J Clin Invest 2004; 113:1498-510. [PMID: 15146248 PMCID: PMC406527 DOI: 10.1172/jci20312] [Citation(s) in RCA: 47] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2003] [Accepted: 03/16/2004] [Indexed: 11/17/2022] Open
Abstract
The clonotypic surface Ig receptor expressed by malignant B cells, idiotype, is a tumor-specific antigen and an attractive target for active immunotherapy. While Ab's specific for tumor idiotype have been well described in patients with B cell malignancies, the precise antigenic epitopes in human idiotype recognized by autologous T cells remain largely unknown. We report here that T cell lines generated from lymphoma patients actively immunized with idiotype protein specifically recognized multiple, unique immunodominant epitopes in autologous tumor idiotype. Synthetic peptides corresponding to hypervariable, but not framework, regions of Ig heavy chain specifically stimulated CD4(+) and CD8(+) T cells to proliferate and secrete proinflammatory cytokines in an MHC-associated manner. Detailed analysis revealed a minimal determinant of an immunodominant epitope, comprising critical residues at the amino terminus that may be a product of somatic hypermutation. Association of idiotype-specific T cell responses with previously documented molecular remissions in idiotype-vaccinated patients suggests that the newly identified T cell epitopes may be clinically relevant. Such antigenic epitopes may serve as candidates for novel peptide-vaccine strategies, and as tools to selectively expand tumor antigen-specific T cells for adoptive immunotherapy and for monitoring T cell immunity in vaccinated patients.
Collapse
Affiliation(s)
- Sivasubramanian Baskar
- Basic Research Program, SAIC-Frederick Inc., National Cancer Institute at Frederick, National Institutes of Health, Maryland, 21702, USA.
| | | | | |
Collapse
|
265
|
Rasmussen T, Knudsen LM, Huynh TK, Johnsen HE. Molecular and clinical follow-up after treatment of multiple myeloma. Acta Haematol 2004; 112:105-10. [PMID: 15179010 DOI: 10.1159/000077565] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
Multiple myeloma (MM) is a B cell malignancy characterized by accumulation of plasma cells (PCs) in the bone marrow. Traditional methods for the detection of minimal residual disease (MRD) measure the presence of monoclonal immunoglobulin protein secreted by the malignant PCs. However, changes in the level of MRD in MM may span 6 logs, and methods with a high sensitivity and dynamic range are necessary for quantitating MRD in MM. The two main technologies used in MRD detection are flow cytometry and patient-specific reverse transcription (RT) PCR. Patient-specific RT-PCR has high sensitivity and may be beneficial in monitoring patients receiving allogeneic transplantation. However, for the MRD evaluation of autotransplants, where few patients achieve molecular remission, flow cytometry monitoring seems to be sufficient.
Collapse
Affiliation(s)
- Thomas Rasmussen
- Department of Hematology L 54P4, Herlev Hospital, University of Copenhagen, Herlev, Denmark.
| | | | | | | |
Collapse
|
266
|
Abstract
The chemokine/chemokine receptor network is an essential part of an intricate system of immunosurveillance and homeostasis, it promotes or suppresses neovascularization, affects and regulates directly or indirectly growth and metastasis of malignant cells. Numerous studies have been conducted to harness this network as therapeutic agents for cancer to redress the chemokine balance and control angiogenesis and tumour growth and metastasis. Second generation of immunotherapeutics and chemoattractant-based vaccines use chemokines and chemoattractant peptides to elicit antitumor immunity by a specific targeting and modulating subsets of effector leukocytes, including professional antigen presenting cells.
Collapse
Affiliation(s)
- Marta Coscia
- Laboratorio di Ematologia Oncologica, Divisione di Ematologia dell'Universita' di Torino, CeRMS, Azienda Ospedaliera San Giovanni Battista, Torino, Italy
| | | |
Collapse
|
267
|
Stevenson FK, Rice J, Ottensmeier CH, Thirdborough SM, Zhu D. DNA fusion gene vaccines against cancer: from the laboratory to the clinic. Immunol Rev 2004; 199:156-80. [PMID: 15233733 DOI: 10.1111/j.0105-2896.2004.00145.x] [Citation(s) in RCA: 67] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
Vaccination against target antigens expressed by cancer cells has now become a realistic goal. DNA vaccines provide a direct link between identification of genetic markers in tumors and vaccine formulation. Simplicity of manufacture facilitates construction of vaccines against disease subsets or even for individual patients. To engage an immune system that exists to fight pathogens, we have developed fusion gene vaccines encoding tumor antigens fused to pathogen-derived sequences. This strategy activates high levels of T-cell help, the key to induction and maintenance of effective immunity. We have dissected the immunogenic tetanus toxin to obtain specific sequences able to activate antibody, CD4+, or CD8+ T cells to attack selected fused tumor antigens. Principles established in preclinical models are now being tested in patients. So far, objective immune responses against idiotypic antigen of neoplastic B cells have been observed in patients with B-cell malignancies and in normal transplant donors. These responses provide a platform for testing physical methods to improve DNA delivery and strategies to boost responses. For cancer, demands are high, because vaccines have to activate powerful immunity against weak antigens, often in a setting of immune damage or tolerance. Vaccination strategies against cancer and against microbes are sharing knowledge and technology for mutual benefit.
Collapse
Affiliation(s)
- Freda K Stevenson
- Molecular Immunology Group, Tenovus Laboratory, Cancer Sciences Division, Southampton University Hospitals, Southampton, UK.
| | | | | | | | | |
Collapse
|
268
|
Orsini E, Pasquale A, Maggio R, Calabrese E, Mauro FR, Giammartini E, Guarini A, Foa R. Phenotypic and functional characterization of monocyte-derived dendritic cells in chronic lymphocytic leukaemia patients: influence of neoplastic CD19+
cells in vivo
and in vitro. Br J Haematol 2004; 125:720-8. [PMID: 15180861 DOI: 10.1111/j.1365-2141.2004.04971.x] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
Dendritic cells (DCs) are the most potent antigen-presenting cells and are therefore an attractive option as antigen carriers in vaccination protocols. Chronic lymphocytic leukaemia (CLL) represents a potential good target for these approaches. The present study was designed to investigate the feasibility of generating in vitro fully functional DCs from peripheral blood (PB) monocytes of CLL patients at different phases of the disease. Although functional DCs could be obtained from CLL samples, in patients with active disease the expression of some co-stimulatory molecules appeared to be reduced. In contrast, DCs from CLL patients in remission showed no difference from those of normal controls. Moreover, patients with active disease produced DCs with reduced allostimulatory ability when compared with normal ones, whereas the functional capacities appeared to be restored in CLL DCs from remission patients. To more precisely assess the possible inhibitory effect of CLL cells on DC development, the influence of autologous leukaemic CD19(+) cells on the generation of monocyte-derived CLL DCs in vitro was investigated. The addition of CLL neoplastic cells markedly affected monocyte-derived DC maturation. In conclusion, monocytes from CLL patients with active disease give rise to DCs, which show phenotypic and functional defects that are not observed in remission CLL patients. These results need to be taken into account in the design of DC-based immunotherapeutic approaches in CLL.
Collapse
MESH Headings
- Adult
- Aged
- Alemtuzumab
- Antibodies, Monoclonal/therapeutic use
- Antibodies, Monoclonal, Humanized
- Antibodies, Monoclonal, Murine-Derived
- Antibodies, Neoplasm/therapeutic use
- Antigens, CD19/immunology
- Antineoplastic Agents/therapeutic use
- Biomarkers/analysis
- Cancer Vaccines
- Case-Control Studies
- Clone Cells
- Coculture Techniques
- Dendritic Cells/immunology
- Female
- Flow Cytometry
- Humans
- Immunotherapy, Adoptive
- Leukemia, Lymphocytic, Chronic, B-Cell/immunology
- Leukemia, Lymphocytic, Chronic, B-Cell/therapy
- Male
- Middle Aged
- Rituximab
Collapse
Affiliation(s)
- Enrica Orsini
- Dipartimento di Biotecnologie Cellulari ed Ematologia, University La Sapienza, Rome, Italy.
| | | | | | | | | | | | | | | |
Collapse
|
269
|
Brown R, Murray A, Pope B, Sze DM, Gibson J, Ho PJ, Hart D, Joshua D. Either interleukin-12 or interferon-γ can correct the dendritic cell defect induced by transforming growth factor β
1
in patients with myeloma. Br J Haematol 2004; 125:743-8. [PMID: 15180863 DOI: 10.1111/j.1365-2141.2004.04984.x] [Citation(s) in RCA: 49] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022]
Abstract
The poor response to immunotherapy in patients with multiple myeloma (MM) indicates that a better understanding of any defects in the immune response in these patients is required before effective therapeutic strategies can be developed. Recently we reported that high potency (CMRF44(+)) dendritic cells (DC) in the peripheral blood of patients with MM failed to significantly up-regulate the expression of the B7 co-stimulatory molecules, CD80 and CD86, in response to an appropriate signal from soluble trimeric human CD40 ligand. This defect was caused by transforming growth factor beta(1) (TGFbeta(1)) and interleukin (IL)-10, produced by malignant plasma cells, and the defect was neutralized in vitro with anti-TGFbeta(1). As this defect could impact on immunotherapeutic strategies and may be a major cause of the failure of recent trials, it was important to identify a more clinically useful agent that could correct the defect in vivo. In this study of 59 MM patients, the relative and absolute numbers of blood DC were only significantly decreased in patients with stage III disease and CD80 up-regulation was reduced in both stage I and stage III. It was demonstrated that both IL-12 and interferon-gamma neutralized the failure to stimulate CD80 up-regulation by huCD40LT in vitro. IL-12 did not cause a change in the distribution of DC subsets that were predominantly myeloid (CD11c+ and CDw123-) suggesting that there would be a predominantly T-helper cell type response. The addition of IL-12 or interferon-gamma to future immunotherapy trials involving these patients should be considered.
Collapse
Affiliation(s)
- Ross Brown
- Institute of Haematology, Royal Prince Alfred Hospital, Sydney, Australia.
| | | | | | | | | | | | | | | |
Collapse
|
270
|
Zhang M, Dong Z, Li J, Zhao G, Chen C, Li A. Humoral immunoreaction induced by TCR DNA vaccine for β chain of T cell lymphoma. Vaccine 2004; 22:2031-41. [PMID: 15121317 DOI: 10.1016/j.vaccine.2003.05.003] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2002] [Revised: 01/14/2003] [Accepted: 05/13/2003] [Indexed: 11/16/2022]
Abstract
We exploited the humoral immunoreaction of mice induced by TCR DNA vaccine of beta chain of T cell lymphoma. The plasmids of pcDNA3.1/TCR V beta 8 was constructed. The BALB/c mice were randomly divided into four groups which were pcDNA3.1, pcDNA3.1/TCR V beta 8, pcDNA3.1/TCR V beta 8 + CpG + liposome and phosphorothioate CpG groups with six mice in each group. Vaccines were injected in bilateral musculus quadriceps femoris of mice in the 0, second, and fourth week, respectively. The antibody formation was tested by indirect immuofluorescence in the 0, second, fourth, sixth and eighth weeks, respectively, before and after immunization. Production of antibody against TCR V beta 8 antigen was observed in the groups of pcDNA3.1/TCR V beta 8 and pcDNA3.1/TCR V beta 8 + CpG + liposome. The antibody titer began to rise in the fourth week and attain the maximal value in the sixth week. The antibody titer in the group of pcDNA3.1/TCR V beta 8 + CpG + liposome was higher than that in the group of pcDNA3.1/TCR V beta 8 in the fourth and eighth weeks (both P<0.01); the antibody titer in the group of pcDNA3.1/TCR V beta 8 + CpG + liposome was markedly higher than that in the group of pcDNA3.1/TCR V beta 8 in the sixth week (P<0.001). The result indicate that TCR V beta 8 antigen can induce formation of special antibody in mice. CpG and liposome can improve the humoral immunoreaction induced by TCR V beta 8 gene vaccine.
Collapse
MESH Headings
- Amino Acid Sequence
- Animals
- Antibody Formation/immunology
- Antibody Specificity
- Base Sequence
- Biolistics
- CpG Islands
- DNA, Complementary/biosynthesis
- DNA, Complementary/genetics
- Endonucleases/immunology
- Fluorescent Antibody Technique
- Humans
- Jurkat Cells
- Lymphoma, T-Cell/immunology
- Male
- Mice
- Mice, Inbred BALB C
- Molecular Sequence Data
- Plasmids/genetics
- Plasmids/immunology
- RNA/analysis
- RNA/biosynthesis
- Receptors, Antigen, T-Cell/genetics
- Receptors, Antigen, T-Cell/immunology
- Reverse Transcriptase Polymerase Chain Reaction
- Transcription, Genetic
- Vaccines, DNA/immunology
Collapse
Affiliation(s)
- Mingzhi Zhang
- Tumor Department, The First Affiliated Hospital, Zhengzhou University, Zhengzhou 450052, China.
| | | | | | | | | | | |
Collapse
|
271
|
Affiliation(s)
- M Bendandi
- Department of Hematology and Cell Therapy Area, University Clinic, University of Navarra, Avda. Pio XII 36, 31008 Pamplona, Spain.
| | | | | |
Collapse
|
272
|
Affiliation(s)
- Thomas S. Kupper
- Department of Dermatology, Brigham and Women's Hospital, Harvard Institutes of Medicine, 77 Avenue Louis Pasteur, Boston, 02115 Massachusetts USA
| | - Robert C. Fuhlbrigge
- Department of Dermatology, Brigham and Women's Hospital, Harvard Institutes of Medicine, 77 Avenue Louis Pasteur, Boston, 02115 Massachusetts USA
| |
Collapse
|
273
|
Neidhart J, Allen KO, Barlow DL, Carpenter M, Shaw DR, Triozzi PL, Conry RM. Immunization of colorectal cancer patients with recombinant baculovirus-derived KSA (Ep-CAM) formulated with monophosphoryl lipid A in liposomal emulsion, with and without granulocyte-macrophage colony-stimulating factor. Vaccine 2004; 22:773-80. [PMID: 14741172 DOI: 10.1016/j.vaccine.2003.08.021] [Citation(s) in RCA: 43] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022]
Abstract
KSA (Ep-CAM) is highly expressed by colorectal cancers. The safety and immunologic effects of a vaccine consisting of recombinant baculovirus-derived KSA formulated with monophosphoryl lipid A (MPL) in liposomes and emulsified in mineral oil were evaluated, with and without co-administration of granulocyte-macrophage colony-stimulating factor (GM-CSF). Eleven patients with metastatic colorectal cancer received three subcutaneous (s.c.) injections of the vaccine at 4-week intervals. Six patients were randomized to also receive human recombinant GM-CSF (rGM-CSF) by subcutaneous injection daily for 4 days with each vaccination. Immunizations with and without rGM-CSF were well tolerated. Seven of the 11 patients developed significant KSA-specific cellular immune responses as assessed by lymphoproliferation and interferon-gamma (IFN-gamma) ELISPOT assays. All nine tested patients developed positive delayed type hypersensitivity reactions. Eight of the 11 patients developed KSA-specific antibody responses. The highest levels of cellular immune responses were observed in patients who received GM-CSF. Immunization with baculovirus-derived KSA formulated with monophosphoryl lipid A in liposomal emulsion is safe and can elicit KSA-specific immune responses. Co-administration of GM-CSF with this formulation is an effective method of generating KSA-specific T-helper (Th) 1-associated cellular immune responses.
Collapse
Affiliation(s)
- Jeffrey Neidhart
- The University of Alabama at Birmingham, WTI 275-J2, 1530 Third Avenue South, Birmingham, AL 35294-3300, USA
| | | | | | | | | | | | | |
Collapse
|
274
|
Mapara MY, Sykes M. Tolerance and cancer: mechanisms of tumor evasion and strategies for breaking tolerance. J Clin Oncol 2004; 22:1136-51. [PMID: 15020616 DOI: 10.1200/jco.2004.10.041] [Citation(s) in RCA: 414] [Impact Index Per Article: 20.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
The development of malignant disease might be seen as a failure of immune surveillance. However, not all tumors are naturally immunogenic, and even among those that are immunogenic, the uncontrolled rapid growth of a tumor may sometimes out-run a robust immune response. Nevertheless, recent evidence suggests that mechanisms of tolerance that normally exist to prevent autoimmune disease may also preclude the development of an adequate antitumor response and that tumors themselves have the ability to thwart the development of effective immune responses against their antigens. A major challenge has been to develop approaches to breaking this tolerance in tumor-bearing hosts, and recent advances in our understanding of antigen presentation and tolerance have led to some promising strategies. An alternative approach is to use T cells from nontumor-bearing, allogeneic hosts in the form of lymphocyte infusions, with or without hematopoietic cell transplantation. Immunotherapy may occur in this setting via the response of nontolerant, tumor antigen-specific T cells from nontumor-bearing hosts or via the powerful destructive effect of an alloresponse directed against antigens shared by malignant cells in the recipient. Approaches to exploiting this beneficial effect without the deleterious consequence of graft-versus-host disease in allogeneic hematopoietic cell recipients are discussed.
Collapse
Affiliation(s)
- Markus Y Mapara
- Department of Hematology and Oncology, University Medical Center Charité, Campus Virchow Klinikum, Humboldt University Berlin, Germany
| | | |
Collapse
|
275
|
Raje N, Hideshima T, Davies FE, Chauhan D, Treon SP, Young G, Tai YT, Avigan D, Gong J, Schlossman RL, Richardson P, Kufe DW, Anderson KC. Tumour cell/dendritic cell fusions as a vaccination strategy for multiple myeloma. Br J Haematol 2004; 125:343-52. [PMID: 15086415 DOI: 10.1111/j.1365-2141.2004.04929.x] [Citation(s) in RCA: 63] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
Multiple myeloma (MM) cells express certain tumour-associated antigens (TAAs) that could serve as targets for active-specific immunotherapy. The aim of the present study was to test the MM/dendritic cell (DC) fusion as a vaccination strategy. We fused MM cells with DC to generate fusion cells (FCs) and tested their antigen presenting cell (APC) function in mixed lymphocyte reactions and cytotoxicity assays. First, the HS Sultan and SK0-007 HAT sensitive human MM cell lines and DCs generated from peripheral blood of normal donors were fused in the presence of 50% polyethylene glycol to form FCs. Next, tumour cells freshly isolated from patients were similarly fused with autologous DCs to generate FCs. The FCs demonstrated a biphenotypic profile, confirmed both by flow-cytometry and dual immunofluorescence microscopy. These FCs induced MM-specific cytotoxicity. FCs, but not MM cells or DCs alone, were potent stimulators of autologous patient T cells. More importantly, FC-primed autologous peripheral blood mononuclear cells demonstrated major histocompatibility complex-restricted MM-specific cytolysis. These studies therefore demonstrated that MM/DC FC can trigger an autologous immune response to MM cells and formed the framework for a clinical trial currently underway.
Collapse
Affiliation(s)
- Noopur Raje
- Jerome Lipper Multiple Myeloma Center, Department of Adult Oncology, Dana-Farber Cancer Institute, Boston, MA 02215, USA
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
276
|
Schultze JL, Fiore F, von Bergwelt-Baildon M. DCs in lymphoma — biology and therapeutic aspects. Cytotherapy 2004; 6:138-47. [PMID: 15203990 DOI: 10.1080/14653240410006095] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Affiliation(s)
- J L Schultze
- Molecular Tumor Biology and Tumor Immunology, Internal Medicine I, Hematology and Oncology Center for Molecular Medicine Cologne, University of Cologne, Cologne, Germany
| | | | | |
Collapse
|
277
|
Lou Q, Kelleher RJ, Sette A, Loyall J, Southwood S, Bankert RB, Bernstein SH. Germ line tumor-associated immunoglobulin VH region peptides provoke a tumor-specific immune response without altering the response potential of normal B cells. Blood 2004; 104:752-9. [PMID: 15054043 DOI: 10.1182/blood-2004-01-0105] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Previous studies have suggested that murine T cells are tolerant to epitopes derived from germ line variable regions of immunoglobulin (Ig) heavy (VH) or light chains. This has lead to the prediction that germ line VH-region epitopes found in neoplastic B cells cannot be used to provoke an antitumor immune response. To test these assumptions and address the question of how such a vaccine may alter the normal B-cell response, an antibody-forming B-cell hybridoma (1H6) expressing a conserved germ line VH gene with specificity for dextran was generated and used as a tumor model. Using algorithms for predicting major histocompatibility complex (MHC) binding, potential MHC class I and II binding peptides were identified within the 1H6 VH region, synthesized, and tested for MHC binding and immunogenicity. We show that germ line VH peptides, when presented by dendritic cells, are immunogenic in vitro and provoke a tumor-specific protective immune response in vivo. We conclude that (1) it is possible to induce a T-cell response to germ line VH peptides; (2) such peptides can be used to generate a B-cell tumor-specific vaccine; and (3) a vaccine targeting VH peptides expressed by the dominant dextran-specific B-cell clonotype had no effect upon the magnitude of the normal B-cell response to dextran.
Collapse
Affiliation(s)
- Qiang Lou
- Department of Microbiology and Immunology, Witebsky Center for Microbial Pathogenesis and Immunology, State University of New York at Buffalo, USA
| | | | | | | | | | | | | |
Collapse
|
278
|
Malyguine A, Strobl SL, Shafer-Weaver KA, Ulderich T, Troke A, Baseler M, Kwak LW, Neelapu SS. A modified human ELISPOT assay to detect specific responses to primary tumor cell targets. J Transl Med 2004; 2:9. [PMID: 15050026 PMCID: PMC415560 DOI: 10.1186/1479-5876-2-9] [Citation(s) in RCA: 31] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2003] [Accepted: 03/29/2004] [Indexed: 11/10/2022] Open
Abstract
Background The desired outcome of cancer vaccination is to induce a potent T cell response which can specifically recognize and eliminate autologous tumor cells in vivo. Accordingly, immunological assays that demonstrate recognition of native tumor cells (tumor-specific) may be more clinically relevant than assays that demonstrate recognition of tumor protein or peptide (antigen-specific). Methods Towards this goal, we adapted the IFN-γ ELISPOT assay to measure immune responses against autologous primary tumor cells in vaccinated cancer patients. As a model system to develop the assay, we utilized peripheral blood mononuclear cells (PBMC) directly isolated from follicular lymphoma patients vaccinated with tumor-derived idiotype protein. Results After optimizing several variables, we demonstrated that the modified IFN-γ ELISPOT assay could be used to reliably and reproducibly determine the tumor-reactive T cell frequency in the PBMC of these patients. The precursor frequency of tumor-reactive T cells was significantly higher in the postvaccine PBMC, compared with prevaccine samples in all patients tested. Furthermore, the specificity of these T cells was established by the lack of reactivity against autologous normal B cells. Conclusions These results demonstrate the feasibility of quantitating tumor-specific T cell responses when autologous, primary tumor cells are available as targets.
Collapse
Affiliation(s)
- Anatoli Malyguine
- Laboratory of Cell-Mediated Immunity, SAIC-Frederick, Inc., National Cancer Institute at Frederick, Frederick, MD 21702 USA
| | - Susan L Strobl
- Laboratory of Cell-Mediated Immunity, SAIC-Frederick, Inc., National Cancer Institute at Frederick, Frederick, MD 21702 USA
| | - Kimberly A Shafer-Weaver
- Laboratory of Cell-Mediated Immunity, SAIC-Frederick, Inc., National Cancer Institute at Frederick, Frederick, MD 21702 USA
| | - Tracy Ulderich
- Laboratory of Cell-Mediated Immunity, SAIC-Frederick, Inc., National Cancer Institute at Frederick, Frederick, MD 21702 USA
| | - Angela Troke
- Laboratory of Cell-Mediated Immunity, SAIC-Frederick, Inc., National Cancer Institute at Frederick, Frederick, MD 21702 USA
| | - Michael Baseler
- Clinical Services Program, SAIC-Frederick, Inc., National Cancer Institute at Frederick, Frederick, MD 21702 USA
| | - Larry W Kwak
- Experimental Transplantation Immunology Branch, Center for Cancer Research, National Cancer Institute at Frederick, Frederick, MD 21702 USA
| | - Sattva S Neelapu
- Experimental Transplantation Immunology Branch, Center for Cancer Research, National Cancer Institute at Frederick, Frederick, MD 21702 USA
| |
Collapse
|
279
|
Upshaw JD, Callihan TR. Absolute CD4 and CD8 counts and CD4-to-CD8 ratios in eight patients with indolent B-cell chronic lymphocytic leukemia. South Med J 2004; 97:236-9. [PMID: 15043329 DOI: 10.1097/01.smj.0000085747.15986.b6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
BACKGROUND There are some patients with B-cell chronic lymphocytic leukemia who exhibit an extraordinary natural resistance to this malignancy, which lasts for many years. In this study, we report the T-cell subset values and ratios in eight such patients. METHODS Impath (New York, NY) evaluated immunophenotyping by performing flow cytometry. Absolute CD4 and CD8 counts and CD4:CD8 ratios were performed at Memphis Pathology Laboratory, Memphis, Tennessee. RESULTS CD4 and CD8 counts and CD4:CD8 ratios were normal in all eight patients, in contrast to the suppressor cell proliferation and low helper-suppressor ratios that have been previously reported in other patients with B-cell chronic lymphocytic leukemia. CONCLUSION These results require further study to determine their significance. Implications for further study are discussed.
Collapse
Affiliation(s)
- Jefferson D Upshaw
- Memphis Cancer Center, Pathology Group of the Midsouth, Memphis, TN, USA
| | | |
Collapse
|
280
|
Pession A, Tonelli R, Canderan G, Bendandi M, Rosolen A, Basso K, Basso G, Locatelli F, Zabalegui N, Paolucci G. Non-radiolabelled PCR consensus primers and automatic sequencing enable rapid identification of tumor-specific V(H) CDR3 in aggressive B-cell malignancies. Leuk Lymphoma 2004; 44:1597-601. [PMID: 14565664 DOI: 10.3109/10428190309178784] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
Abstract
Even though the results of current therapy are improved for B-cell acute lymphoblastic leukemia (B-ALL) and Burkitt's lymphoma (BL), prognosis of relapsed mature B-ALL and BL still remain extremely poor. In this study, we investigated the possibility of applying the use of non-radiolabelled PCR consensus primers and automatic sequencing for the rapid identification of the tumor-specific V(H) CDR3 nucleotide sequence, in mature B-ALL and BL. RNA was extracted from four consecutive, unselected samples from BL cases and three consecutive, unselected samples from mature B-ALL cases. The feasibility of the identification of the tumor-specific V(H) CDR3 nucleotide sequence was then assessed by using non-radiolabelled PCR consensus primers with automatic sequencing. The tumor-specific V(H) CDR3 nucleotide sequence was successfully identified for all seven patients (3 mature B-ALL and BL). The time required was substantially lower than that of the other methods previously published, despite the poor quality of some of the samples. The procedure showed rapidity, reliability and reproducibility. The characteristics of the methodology applied widen the possibility of developing anti-idiotypic therapeutic strategies, even in these B-cell malignancies.
Collapse
Affiliation(s)
- Andrea Pession
- Department of Pediatrics, University of Bologna, Ospedale S. Orsola, Via Massarenti 11, 40100 Bologna, Italy.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
281
|
|
282
|
Coscia M, Mariani S, Battaglio S, Di Bello C, Fiore F, Foglietta M, Pileri A, Boccadoro M, Massaia M. Long-term follow-up of idiotype vaccination in human myeloma as a maintenance therapy after high-dose chemotherapy. Leukemia 2004; 18:139-45. [PMID: 14574332 DOI: 10.1038/sj.leu.2403181] [Citation(s) in RCA: 50] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
The aim of this work was to evaluate the long-term immunological and clinical impact of idiotype (Id) vaccination in multiple myeloma (MM) patients in first remission after high-dose chemotherapy. A total of 15 patients received a series of subcutaneous (s.c.) injections of autologous Id, conjugated to keyhole limpet hemocyanin (KLH) and in association with low doses of GM-CSF. The median duration of follow-up was 110 months from diagnosis. The vaccine induced immune responses that lasted almost 2 years after the end of treatment. Antibody responses included anti-KLH IgM and IgG (90% of patients), anti-KLH IgE (30%), anti-GM-CSF IgG (20%), anti-Id IgG (20%), and anti-Id IgE (30%). Id-specific delayed type hypersensitivity skin tests were positive in 85% of tested patients. Following vaccination, a progressive recovery of T-cell receptor (TCR) diversity was observed and the loss of oligoclonality was significantly correlated with the remission duration. Although Id/KLH conjugates did not eliminate the residual tumor burden, the median progression-free survival, and overall survival were 40 and 82 months, respectively. A retrospective case-matched analysis showed similar results in patients treated with IFN-alpha alone or in association with steroids. This vaccine formulation can overcome Id-specific immune tolerance by inducing clinical responses that are worthy of further investigation.
Collapse
MESH Headings
- Adjuvants, Immunologic/administration & dosage
- Antibodies, Anti-Idiotypic/immunology
- Antibodies, Anti-Idiotypic/metabolism
- Antibodies, Neoplasm/immunology
- Antibodies, Neoplasm/metabolism
- Antineoplastic Combined Chemotherapy Protocols/administration & dosage
- Case-Control Studies
- Combined Modality Therapy
- Follow-Up Studies
- Glucocorticoids/administration & dosage
- Granulocyte-Macrophage Colony-Stimulating Factor/administration & dosage
- Hematopoietic Stem Cell Transplantation
- Hemocyanins/administration & dosage
- Humans
- Hypersensitivity, Delayed/immunology
- Immunity, Cellular
- Immunoglobulin Idiotypes/immunology
- Immunoglobulin Idiotypes/therapeutic use
- Injections, Subcutaneous
- Interferon-alpha/administration & dosage
- Middle Aged
- Multiple Myeloma/immunology
- Multiple Myeloma/prevention & control
- Multiple Myeloma/therapy
- Neoplasm Staging
- Receptors, Antigen, T-Cell/metabolism
- Remission Induction
- Retrospective Studies
- Survival Rate
- Treatment Outcome
- Vaccination
Collapse
Affiliation(s)
- M Coscia
- Centro di Ricerca in Medicina Sperimentale, Ospedale San Giovanni Battista, Torino, Italy
| | | | | | | | | | | | | | | | | |
Collapse
|
283
|
Sabel MS, Skitzki J, Stoolman L, Egilmez NK, Mathiowitz E, Bailey N, Chang WJ, Chang AE. Intratumoral IL-12 and TNF-α–Loaded Microspheres Lead To Regression of Breast Cancer and Systemic Antitumor Immunity. Ann Surg Oncol 2004; 11:147-56. [PMID: 14761917 DOI: 10.1245/aso.2004.03.022] [Citation(s) in RCA: 52] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022]
Abstract
BACKGROUND Local, sustained delivery of cytokines at a tumor can enhance induction of antitumor immunity and may be a feasible neoadjuvant immunotherapy for breast cancer. We evaluated the ability of intratumoral poly-lactic-acid-encapsulated microspheres (PLAM) containing interleukin 12 (IL-12), tumor necrosis factor alpha (TNF-alpha), and granulocyte-macrophage colony stimulating factor (GM-CSF) in a murine model of breast cancer to generate a specific antitumor response. METHODS BALB/c mice with established MT-901 tumors underwent resection or treatment with a single intratumoral injection of PLAM containing IL-12, TNF-alpha, or GM-CSF, alone or in combination. Two weeks later, lymph nodes and spleens were harvested, activated with anti-CD3 monoclonal antibodies (mAb) and rhIL-2, and assessed for antitumor reactivity by an interferon gamma (IFNgamma) release assay. Tumor-infiltrating lymphocyte (TIL) analysis was performed on days 2 and 5 after treatment by mechanically processing the tumors to create a single cell suspension, followed by three-color fluorescence-activated cell sorter (FACS) analysis. RESULTS Intratumoral injection of cytokine-loaded PLAM significantly suppressed tumor growth, with the combination of IL-12 and TNF-alpha leading to increased infiltration by polymorphonuclear cells and CD8+ T-cells in comparison with controls. The induction of tumor-specific reactive T-cells in the nodes and spleens, as measured by IFN-gamma production, was highest with IL-12 and TNF-alpha. This treatment resulted in resistance to tumor rechallenge. CONCLUSIONS A single intratumoral injection of IL-12 and TNF-alpha-loaded PLAM into a breast tumor leads to infiltration by polymorphonuclear cells and CD8+ T-cells with subsequent tumor regression. In addition, this local therapy induces specific antitumor T-cells in the lymph nodes and spleens, resulting in memory immune response.
Collapse
Affiliation(s)
- Michael S Sabel
- Division of Surgical Oncology, University of Michigan, Ann Arbor, Michigan, USA.
| | | | | | | | | | | | | | | |
Collapse
|
284
|
Kwak LW, Neelapu SS, Bishop MR. Adoptive immunotherapy with antigen-specific T cells in myeloma: a model of tumor-specific donor lymphocyte infusion. Semin Oncol 2004; 31:37-46. [PMID: 14970936 DOI: 10.1053/j.seminoncol.2003.11.001] [Citation(s) in RCA: 17] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Abstract
Although partial remissions rates of up to 60% are obtained with conventional therapeutic regimens, multiple myeloma is essentially an incurable disease with a median survival of approximately 30 months. Allogeneic stem cell transplantation (SCT) results in a high percentage of complete remissions (CRs), but it can be associated with significant treatment-related mortality. Recent clinical studies have shown that highly immunosuppressive, yet nonmyeloablative, doses of fludarabine-based chemotherapy can result in alloengraftment. However, even with a reduction in treatment-related mortality, success with allogeneic SCT is limited by the significant risk of relapse. The goal of the strategy described is to transfer tumor antigen-specific immunity induced in the stem cell donor to the allogeneic SCT recipient to reduce relapse. Donors are immunized with a well-defined vaccine, specific for the patient's tumor. The allogeneic SCT is performed with a conditioning regimen consisting of cyclophosphamide and fludarabine, and the stem cell source is blood mobilized with filgrastim, which could potentially enhance the transfer of a larger number of tumor-specific T cells in the allograft, as compared to bone marrow. Donor immunization with myeloma idiotype protein in the setting of a nonmyeloablative SCT may represent a novel strategy for the treatment of myeloma.
Collapse
Affiliation(s)
- Larry W Kwak
- Experimental Transplantation and Immunology Branch, Center for Cancer Research, National Cancer Institute, NIH, Bethesda, MD 20892, USA
| | | | | |
Collapse
|
285
|
Takedatsu H, Okamura T, Yoshimoto K, Harada M, Koga M, Shichijo S, Sata M, Itoh K. Expression of Epithelial Cancer-Related Antigens in Hematologic Malignancies Applicable for Peptide-Based Immunotherapy. J Immunother 2004; 27:289-97. [PMID: 15235390 DOI: 10.1097/00002371-200407000-00005] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
Recent advances in tumor immunology have resulted in identification of many epithelial cancer-related antigens and peptides applicable to specific immunotherapy. The authors investigated whether these peptides, which are being studied clinically, could be appropriate target molecules for treatment of patients with hematologic malignancies. The majority of hematologic malignant cells studied expressed five different epithelial cancer-related antigens. Cytotoxic T lymphocyte (CTL) precursors reactive to these antigen-derived peptides were detected in peripheral blood mononuclear cells (PBMCs) of the majority of HLA-A24 patients, and the mean number of peptides recognized by CTL precursors was 2.4 per patient, ranging from 0 to 8 among the 10 peptides tested. These peptide-stimulated PBMCs exhibited HLA-A24-restricted cytotoxic activity against hematologic malignant cells but not against blastoid T cells. More importantly, these peptide-stimulated PBMCs exhibited cytotoxicity against freshly prepared autologous malignant cells in an HLA-A24-restricted manner. These results may provide a scientific basis for the use of these peptides from epithelial cancer-related antigens in specific immunotherapy for patients with hematologic malignancies.
Collapse
Affiliation(s)
- Hiroko Takedatsu
- Department of Immunology, Kurume University School of Medicine, Kurume, Japan.
| | | | | | | | | | | | | | | |
Collapse
|
286
|
Costello RT, Fauriat C, Rey J, Gastaut JA, Olive D. Immunobiology of haematological malignant disorders: the basis for novel immunotherapy protocols. Lancet Oncol 2004; 5:47-55. [PMID: 14700608 DOI: 10.1016/s1470-2045(03)01323-8] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
The immune system is a complex arrangement of cellular interactions that preserve the integrity of a organism by elimination of all elements judged dangerous. However, the development of tumours in immunocompetent patients suggests the existence of an imbalance that favours tumour cells against the immune response. What are the different possibilities for reversing this process to drive an efficient antitumour response? We discuss, focusing on the haematological features, classic immunity (ie, antigen-specific and HLA-restricted immunity). We address the central issues of tumour antigen presentation and recognition and their possible clinical use. Last, we discuss non-HLA-restricted immunity, which does not require the recognition of specific antigens and relies on particular cell populations such as natural killer cells.
Collapse
Affiliation(s)
- Régis T Costello
- Institut Paoli-Calmettes, Université de la Méditerranée, Marseille, France
| | | | | | | | | |
Collapse
|
287
|
Timmerman JM. Therapeutic idiotype vaccines for non-Hodgkin's lymphoma. ADVANCES IN PHARMACOLOGY (SAN DIEGO, CALIF.) 2004; 51:271-93. [PMID: 15464914 DOI: 10.1016/s1054-3589(04)51012-8] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/10/2022]
Affiliation(s)
- John M Timmerman
- Division of Hematology/Oncology, University of California, Los Angeles Center for Health Sciences, 42-121 Los Angeles, California 90095-1678, USA
| |
Collapse
|
288
|
Affiliation(s)
- Freda K Stevenson
- Molecular Immunology Group, Tenovus Laboratory, Cancer Sciences Division Southampton University Hospitals Trust, Southampton SO16 6YD, United Kingdom
| | | | | |
Collapse
|
289
|
Caligiuri MA, Velardi A, Scheinberg DA, Borrello IM. Immunotherapeutic Approaches for Hematologic Malignancies. Hematology 2004:337-53. [PMID: 15561691 DOI: 10.1182/asheducation-2004.1.337] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/28/2023] Open
Abstract
Abstract
The immune system has two complementary arms: one is older and seemingly more primitive, called the innate immune system, found in both plants and animals. The second (already many millions of years old!) is the adaptive or antigen-specific immune system, limited to vertebrate animals. The human innate immune system has many cellular elements that include granulocytes, monocytes, macrophages, natural killer (NK) cells, mast cells, eosinophils, and basophils. Receptors for these cells are non-clonal, fixed in the genome, requiring no rearrangement, and recognize conserved molecular patterns that are specific to pathogens. The adaptive immune system (B cells and T cells) have receptors with great variation, able to recognize an almost an unlimited number of highly specific pathogens through rearrangement of receptor gene segments, and can also provide immunological memory so critical for vaccination. As the immune system has evolved to recognize non-self, malignant transformation of self can likely escape immune surveillance with relative ease. Contributors to this chapter are utilizing distinct components of either the innate or adaptive immune system that recognize non-self, in combination with what we know about differences between malignant and normal self, in an effort to develop novel and effective immunologic approaches against hematologic malignancies.
In Section I, Dr. Andrea Velardi reviews the benefits of NK cell alloreactivity in mismatched hematopoietic transplantation, provides updates on current clinical trials, and discusses further therapeutic perspectives emerging from murine bone marrow transplant models.
In Section II, Dr. David Scheinberg reviews novel leukemic antigens being targeted by humanized monoclonal antibodies as well as mechanisms by which antibody-mediated cytotoxicity occurs in vivo.
In Section III, Dr. Ivan Borrello reviews vaccine and adoptive T cell immunotherapy in the treatment of hematologic malignancies. Specifically, he discusses the various vaccine approaches used as well as strategies aimed at augmenting the tumor specificity of T cell therapies.
Collapse
|
290
|
Abstract
Although multiple myeloma remains a terminal illness, the past four decades have seen a dramatic change in the outlook for a newly diagnosed patient in terms of therapies available, supportive care and insight into the pathogenesis of this disease. Among the newer agents available for treatment, thalidomide has been resurrected and discovered to be a valuable therapy for myeloma. Thalidomide appears to work, at least in part, through its anti-angiogenic properties, but much remains to be learned about its mechanism of action as well as optimal administration regimens. With the development of increasingly more potent bisphosphonates it has become possible to diminish the painful skeletal complications of myeloma, one of the most devastating problems of this disease. The most recent generation of bisphosphonates, pamidronic acid and zoledronic acid, have provided a statistically significant decrease in the skeletal complications of myeloma when used in a prophylactic manner. These agents appear to work by inhibiting osteoclast function. Progressive improvement in cytogenetic techniques has now demonstrated that almost all patients with myeloma have chromosomal abnormalities, some of which appear to confer varying degrees of prognostic significance. In particular, the changes in chromosome 13 are associated with an unusually poor outcome. These findings are serving as a guide toward learning more about the pathogenesis of myeloma as well as in identifying potential targets for therapy. Stem cell transplantation has emerged as the standard treatment for the large majority of patients with myeloma following the demonstration of superior complete remission and survival, both disease-free and overall, in a French randomised trial. Unfortunately, virtually all patients will eventually relapse following autologous stem cell transplantation, prompting continuing efforts such as tandem transplants, CD34+ selection, as well as modifications in the conditioning regimen to improve outcomes. Allogeneic bone marrow transplants appear to offer a better chance for a possible cure of myeloma but have been associated with an unusually high mortality. However, this approach is being revived with the advent of the less toxic non-myeloablative transplant that has provided an 81% short-term survival in a trial combining this approach with an initial conventional autologous bone marrow transplant. Immunotherapy with dendritic cells appears now to be a feasible way to enhance innate or acquired immunity to help eliminate minimal residual disease following autologous bone marrow transplant. Unfortunately, a cure for myeloma remains elusive but the continuing advances in management may significantly prolong survival in affected patients.
Collapse
Affiliation(s)
- Leonard T Heffner
- Winship Cancer Institute, Emory University School of Medicine, Atlanta, Georgia 30322, USA.
| | | |
Collapse
|
291
|
Nelson EL, Prieto D, Alexander TG, Pushko P, Lofts LA, Rayner JO, Kamrud KI, Fralish B, Smith JF. Venezuelan Equine Encephalitis Replicon Immunization Overcomes Intrinsic Tolerance and Elicits Effective Anti-tumor Immunity to the ‘Self’ tumor-associated antigen, neu in a Rat Mammary Tumor Model. Breast Cancer Res Treat 2003; 82:169-83. [PMID: 14703064 DOI: 10.1023/b:brea.0000004373.09678.bb] [Citation(s) in RCA: 29] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023]
Abstract
Many tumor-associated antigens (TAAs) represent 'self' antigens and as such, are subject to the constraints of immunologic tolerance. There are significant barriers to eliciting anti-tumor immune responses of sufficient magnitude. We have taken advantage of a Venezuelan equine encephalitis-derived alphavirus replicon vector system with documented in vivo tropism for immune system dendritic cells. We have overcome the intrinsic tolerance to the 'self' TAA rat neu and elicited an effective anti-tumor immune response using this alphavirus replicon vector system and a designed target antigen in a rigorous rat mammary tumor model. We have demonstrated the capacity to generate 50% protection in tumor challenge experiments (p = 0.004) and we have confirmed the establishment of immunologic memory by both second tumor challenge and Winn Assay (p = 0.009). Minor antibody responses were identified and supported the establishment of T helper type 1 (Th1) anti-tumor immune responses by isotype. Animals surviving in excess of 300 days with established effective anti-tumor immunity showed no signs of autoimmune phenomena. Together these experiments support the establishment of T lymphocyte dependent, Th1-biased anti-tumor immune responses to a non-mutated 'self' TAA in an aggressive tumor model. Importantly, this tumor model is subject to the constraints of immunologic tolerance present in animals with normal developmental, temporal, and anatomical expression of a non-mutated TAA. These data support the continued development and potential clinical application of this alphaviral replicon vector system and the use of appropriately designed target antigen sequences for anti-tumor immunotherapy.
Collapse
MESH Headings
- Amino Acid Sequence
- Animals
- Antigens, Neoplasm/immunology
- Disease Models, Animal
- Encephalitis Virus, Venezuelan Equine/genetics
- Female
- Genetic Vectors/immunology
- Genetic Vectors/therapeutic use
- Humans
- Immunization
- Mammary Neoplasms, Experimental/immunology
- Mammary Neoplasms, Experimental/therapy
- Molecular Sequence Data
- Neoplasm Proteins/immunology
- Rats
- Rats, Inbred F344
- Receptor, ErbB-2/chemistry
- Receptor, ErbB-2/genetics
- Receptor, ErbB-2/immunology
- Replicon/immunology
- Self Tolerance
- Vaccines, Synthetic/immunology
Collapse
Affiliation(s)
- Edward L Nelson
- Department of Medicine, Division of Hematology/Oncology, School of Medicine and Molecular Biology and Biochemistry, University of California, Irvine, CA 97697-4060, USA.
| | | | | | | | | | | | | | | | | |
Collapse
|
292
|
Affiliation(s)
- Helen McCarthy
- Tenovus Laboratory, Cancer Sciences Division, Southampton University Hospitals Trust, Southampton, UK
| | | | | | | |
Collapse
|
293
|
Laport GG, Levine BL, Stadtmauer EA, Schuster SJ, Luger SM, Grupp S, Bunin N, Strobl FJ, Cotte J, Zheng Z, Gregson B, Rivers P, Vonderheide RH, Liebowitz DN, Porter DL, June CH. Adoptive transfer of costimulated T cells induces lymphocytosis in patients with relapsed/refractory non-Hodgkin lymphoma following CD34+-selected hematopoietic cell transplantation. Blood 2003; 102:2004-13. [PMID: 12763934 DOI: 10.1182/blood-2003-01-0095] [Citation(s) in RCA: 166] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023] Open
Abstract
We explored the feasibility and toxicity of administering escalating doses of anti-CD3/CD28 ex vivo costimulated T cells as a therapeutic adjunct for patients with relapsed, refractory, or chemotherapy-resistant, aggressive non-Hodgkin lymphoma (NHL) following high-dose chemotherapy and CD34+-selected hematopoietic cell transplantation (HCT). Sixteen patients had infusions on day 14 after HCT of autologous T cells that had been stimulated using beads coated with anti-CD3 and anti-CD28 monoclonal antibodies. At baseline, the subjects had severe quantitative and functional T-cell impairments. The culture procedure partially reversed impaired cytokine responsiveness in T cells in vitro and in vivo. Transient dose-dependent infusion toxicities were observed. There was a rapid reconstitution of lymphocytes; however, there were persistent defects in CD4 T cells. Most interestingly, 5 patients had a delayed lymphocytosis between day 30 and day 120 after HCT. Maximal clinical responses included 5 patients with a complete response (CR), 7 patients with a partial response (PR), and 4 patients with stable disease. At a median follow-up of 33 months (range, 26-60 months), 5 patients are alive with stable or relapsed disease and 3 patients remain in CR. In conclusion, this phase 1 trial demonstrates that adoptive transfer of autologous costimulated T cells (1) is feasible in heavily pretreated patients with advanced NHL, (2) is associated with a rapid recovery of lymphocyte counts, (3) reverses cytokine activation deficits in vitro, and (4) is associated with delayed lymphocytosis in a subset of patients.
Collapse
Affiliation(s)
- Ginna G Laport
- Translational Research Program, Abramsonm Family Cancer Research Institute, University of Pennsylvania School of Medicine, 421 Curie Blvd, Philadelphia, PA 19104-6160, USA
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
294
|
Ladetto M, Mantoan B, Ricca I, Astolfi M, Drandi D, Compagno M, Vallet S, dell'Aquila M, Alfarano A, Rossatto P, Rocci A, Vitolo U, Corradini P, Boccadoro M, Tarella C. Recurrence of Bcl-2/IgH polymerase chain reaction positivity following a prolonged molecular remission can be unrelated to the original follicular lymphoma clone. Exp Hematol 2003; 31:784-8. [PMID: 12962724 DOI: 10.1016/s0301-472x(03)00191-7] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/27/2022]
Abstract
OBJECTIVE The aim of this study was to evaluate whether reappearance of polymerase chain reaction (PCR) positivity for the Bcl-2/IgH translocation following a phase of molecular remission in autografted follicular lymphoma (FL) patients is always associated with reappearance of the original neoplastic clone. PATIENTS AND METHODS The molecular follow-up of 119 autografted Bcl-2/IgH positive patients was evaluated by nested PCR. In case of molecular recurrence, direct sequencing of involved rearrangements has been performed both at diagnosis and at the time of recurrence. The two sequences then were compared in terms of breakpoints, N insertions, and JH usage. RESULTS Seventy-five patients achieving molecular remission were identified in our patient sample (63%). Of these patients, eight (10.6%) experienced molecular recurrence. Direct sequencing of the Bcl-2/IgH translocation performed at diagnosis and recurrence showed identical rearrangements in six subjects and unrelated rearrangements in two. As opposed to most true molecular relapses, unrelated rearrangements always occurred several years after transplantation. To date, the two subjects carrying unrelated rearrangements show no signs of active lymphoproliferative disease. CONCLUSIONS This report is the first evidence that Bcl-2/IgH rearrangements unrelated to the original tumor clone can lead to false-positive results during the molecular follow-up of autografted FL patients. Based on these results, we recommend confirmation by direct sequencing, at least for patients experiencing molecular relapse 2 or more years after the end of treatment. This will be particularly important for patients enrolled in clinical trials that schedule additional treatment in case of molecular evidence of persistent disease activity.
Collapse
Affiliation(s)
- Marco Ladetto
- Divisione di Ematologia Dipartimento di Medicina ed Oncologia Sperimentale, Università di Torino, A.O. San Giovanni Battista della Citta' di Torino, Torino, Italy.
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
295
|
Winter D, Fiebiger E, Meraner P, Auer H, Brna C, Strohal R, Trautinger F, Knobler R, Fischer GF, Stingl G, Maurer D. Definition of TCR epitopes for CTL-mediated attack of cutaneous T cell lymphoma. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2003; 171:2714-24. [PMID: 12928425 DOI: 10.4049/jimmunol.171.5.2714] [Citation(s) in RCA: 18] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
Therapeutic vaccination against cutaneous T cell lymphoma (CTCL) requires the characterization of cancer cell-specific CTL epitopes. Despite reported evidence for tumor-reactive cytotoxicity in CTCL patients, the nature of the recognized determinants remains elusive. The clonotypic TCR of CTCL cells is a promising candidate tumor-specific Ag. In this study, we report that the clonotypic and framework regions of the TCRs expressed in the malignant T cell clones of six CTCL patients contain multiple peptides with anchor residues fitting the patients' MHC class I molecules. We demonstrate that TCR peptide-specific T cells from the blood of healthy donors and patients can be induced to become cytotoxic effectors after repeated stimulation with 6 of 11 selected peptides with experimentally proven affinity for HLA-A*0201. Importantly, 4 of these 6 CTL lines reproducibly recognize and lyse autologous primary CTCL cells in MHC class I/CD8-dependent fashion. These tumoricidal CTL lines are directed against epitopes from V, hypervariable, and C regions of TCRalpha. We therefore conclude that recombined as well as V framework regions of the tumor cell TCRs contain predictable epitopes for CTL-mediated attack of CTCL cells. Our data further suggest that such peptides represent valuable tools for future anti-CTCL vaccination approaches.
Collapse
MESH Headings
- Amino Acid Sequence
- Antigen Presentation/immunology
- Cancer Vaccines/immunology
- Cancer Vaccines/metabolism
- Cancer Vaccines/therapeutic use
- Cell Line, Tumor
- Clone Cells
- Cytotoxicity, Immunologic/immunology
- Endopeptidases/metabolism
- Epitopes, T-Lymphocyte/blood
- Epitopes, T-Lymphocyte/immunology
- Epitopes, T-Lymphocyte/metabolism
- Epitopes, T-Lymphocyte/therapeutic use
- Histocompatibility Antigens Class I/metabolism
- Humans
- Hydrolysis
- Lymphocyte Activation
- Lymphoma, T-Cell/blood
- Lymphoma, T-Cell/enzymology
- Lymphoma, T-Cell/immunology
- Lymphoma, T-Cell/prevention & control
- Molecular Sequence Data
- Peptide Fragments/blood
- Peptide Fragments/immunology
- Peptide Fragments/metabolism
- Protein Binding/immunology
- Receptors, Antigen, T-Cell/blood
- Receptors, Antigen, T-Cell/immunology
- Receptors, Antigen, T-Cell/metabolism
- Receptors, Antigen, T-Cell/therapeutic use
- Skin Neoplasms/blood
- Skin Neoplasms/enzymology
- Skin Neoplasms/immunology
- Skin Neoplasms/prevention & control
- T-Lymphocytes, Cytotoxic/enzymology
- T-Lymphocytes, Cytotoxic/immunology
- T-Lymphocytes, Cytotoxic/metabolism
Collapse
Affiliation(s)
- Dorian Winter
- Division of Immunology, Allergy, and Infectious Diseases, Department of Dermatology, University of Vienna Medical School, Waehringer Guertel 189-20, A-1090 Vienna, Austria
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
296
|
Abstract
PURPOSE The therapeutic options for treating patients with lymphoma have dramatically expanded with the advent of immune-based treatments. While monoclonal antibodies have quickly become incorporated into the therapeutic armamentarium in the treatment of lymphoma, others have struggled to find a niche. This review will summarize the state of the art of this burgeoning therapy. RECENT FINDINGS The optimal use and timing of existing immunotherapies such as rituximab and radioimmunoconjugates has generated much data in the past few years. Other research has concentrated on the development of newer generation antibodies with alternate binding sites, such as epratuzumab and HU1D10, and bispecific antibodies that can directly interact with cellular immunity. Furthermore, the role of therapeutic vaccines continued to be an important ongoing research question. SUMMARY This review will enumerate the available and most promising developments in immunotherapy as well as provide a working framework to incorporate them into the treatment paradigm for patients with lymphoma.
Collapse
Affiliation(s)
- Jesús G Berdeja
- Loma Linda University & Medical Center, Department of Medicine, California 92354, USA.
| |
Collapse
|
297
|
Abstract
Vaccination strategies for lymphomas were developed along with one of the first recognized tumor-specific targets, the clonal antigen receptor, composed of unique variable regions known as idiotypes. Human clinical trials of idiotype vaccination have benefited from highly concordant animal models, leading to sequential improvements in design. Evidence of the clinical benefit of idiotype vaccines is strong but formally unproven. Significant progress has been made in our understanding of the basic mechanisms underlying the induction of immune responses, which has led to a proliferation of rationally designed immunotherapeutic strategies. Current research efforts include the development of more convenient methods to produce individual idiotype vaccines, the establishment of definitive proof for clinical efficacy, and the implementation of alternative vaccination strategies, including genetic vaccination and genetically or immunologically modified autologous tumor cells and dendritic cells.
Collapse
Affiliation(s)
- Mohammed M Dar
- National Cancer Institute-Frederick, Building 567, Room 203, Frederick, MD 21702-1201, USA
| | | |
Collapse
|
298
|
Hayashi T, Hideshima T, Akiyama M, Raje N, Richardson P, Chauhan D, Anderson KC. Ex vivo induction of multiple myeloma-specific cytotoxic T lymphocytes. Blood 2003; 102:1435-42. [PMID: 12714512 DOI: 10.1182/blood-2002-09-2828] [Citation(s) in RCA: 95] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Multiple myeloma (MM) is an incurable plasma cell malignancy characterized by immunosuppression. In this study, we identified factors in patients' bone marrow (BM) sera inhibiting autologous anti-MM immunity and developed an ex vivo strategy for inducing MM-specific cytotoxic T lymphocytes (CTLs). We found that sera from BM of MM patients inhibited induction of dendritic cells (DCs), evidenced by both phenotype and only weak stimulation of T-cell proliferation. Anti-vascular endothelial growth factor (anti-VEGF) and/or anti-interleukin 6 (anti-IL-6) antibodies neutralized this inhibitory effect, confirming that VEGF and IL-6, at least in part, mediate immunosuppression in MM patients. To induce MM-specific CTLs ex vivo, immature DCs were generated by culture of adherent mononuclear cells in medium containing granulocyte-macrophage colony-stimulating factor (GM-CSF) and IL-4 for 5 days and then cocultured with apoptotic MM bodies in the presence of tumor necrosis factor alpha (TNF-alpha) for 3 days to induce their maturation. Autologous BM or peripheral blood mononuclear cells were stimulated weekly with these DCs, and cytotoxicity was examined against the MM cells used to pulse DCs. DCs cultured with apoptotic bodies stimulated significantly greater T-cell proliferation (stimulation index [SI] = 23.2 at a T-DC ratio of 360:1) than T cells stimulated by MM cells only (SI = 5.6), DCs only (SI = 9.3), or MM lysate-pulsed DCs (SI = 13.5). These CTLs from MM patients demonstrated specific cytotoxicity (24.7% at the effector-target [E/T] ratio of 40:1) against autologous primary MM cells. These studies therefore show that CTLs from MM patients can recognize and lyse autologous tumor cells and provide the framework for novel immunotherapy to improve patient outcome in MM.
Collapse
Affiliation(s)
- Toshiaki Hayashi
- Jerome Lipper Multiple Myeloma Center, Department of Medical Oncology, Dana-Farber Cancer Institute, 44 Binney St, Boston, MA 02115, USA
| | | | | | | | | | | | | |
Collapse
|
299
|
Abstract
Treatment of malignant non-Hodgkin lymphomas (NHL) in the 21st century has been revolutionized by novel biological agents offering targeted approaches in addition to radio-chemotherapy. Monoclonal antibodies (MoAbs) against lymphatic surface antigens have been effective as monotherapeutic agents, and have already shown their superiority to conventional strategies when combined with chemotherapy. Radioimmunoconjugates are more effective than unlabelled antibodies. Specific inhibitors of neoangiogenesis as well as intracellular signal transduction pathways and antiapoptotic mechanisms have shown their efficacy in phase II studies. Long-term improvement in the setting of minimal residual disease has been observed after vaccination against surface antigens and non-myeloablative allogeneic stem cell transplantation is effective in controlling lymphoma growth. Novel specific antigens are currently identified using expression profiling of lymphomas. In the near future, combinations of biological agents will challenge conventional therapy. These exciting new strategies will improve the success rate in aggressive NHLs and may even challenge the paradigm of incurability of indolent lymphomas.
Collapse
MESH Headings
- Alemtuzumab
- Angiogenesis Inhibitors/therapeutic use
- Antibodies, Monoclonal/therapeutic use
- Antibodies, Monoclonal, Humanized
- Antibodies, Monoclonal, Murine-Derived
- Antibodies, Neoplasm/therapeutic use
- Antineoplastic Agents/pharmacology
- Antineoplastic Agents/therapeutic use
- Antineoplastic Combined Chemotherapy Protocols/therapeutic use
- Boronic Acids/therapeutic use
- Bortezomib
- Cancer Vaccines/administration & dosage
- Clinical Trials as Topic
- Clinical Trials, Phase I as Topic
- Clinical Trials, Phase II as Topic
- Clinical Trials, Phase III as Topic
- Cyclophosphamide/therapeutic use
- Disease-Free Survival
- Doxorubicin/therapeutic use
- Forecasting
- Genetic Therapy
- Humans
- Immunoconjugates
- Immunotherapy
- Lymphoma, B-Cell/drug therapy
- Lymphoma, Non-Hodgkin/drug therapy
- Lymphoma, Non-Hodgkin/mortality
- Lymphoma, Non-Hodgkin/radiotherapy
- Lymphoma, Non-Hodgkin/therapy
- Prednisone/therapeutic use
- Protease Inhibitors/therapeutic use
- Proteomics
- Pyrazines/therapeutic use
- Radioimmunotherapy
- Randomized Controlled Trials as Topic
- Rituximab
- Signal Transduction/drug effects
- Survival Analysis
- Thalidomide/therapeutic use
- Time Factors
- Vincristine/therapeutic use
Collapse
Affiliation(s)
- Ulrich Jäger
- Division of Hematology and Hemostaseology, Department of Internal Medicine I, University of Vienna, Vienna, Austria.
| |
Collapse
|
300
|
Abstract
Multiple myeloma (MM) affects 15,000 new patients annually in the US, with 50,000 total patients, and remains incurable. Our preliminary in vitro and animal studies suggest a role for MM-host interactions in regulating MM cell growth, drug resistance, and migration in the bone marrow. Importantly, treatment strategies which target mechanisms whereby MM cells grow and survive in the bone marrow, including thalidomide and its potent immunomodulatory derivatives and proteasome inhibitor PS-341, can overcome classical drug resistance in preclinical and early clinical studies.
Collapse
Affiliation(s)
- Teru Hideshima
- Jerome Lipper Multiple Myeloma Center, Dana-Farber Cancer Institute and Harvard Medical School, Boston, MA 02115, USA
| | | | | |
Collapse
|