Yonsei Med J. 2007 Feb;48(1):1-10. English.
Published online Feb 20, 2007.
Copyright © 2007 The Yonsei University College of Medicine
Review

Therapeutic Vaccine for Lymphoma

Seung-Tae Lee, Sattva S. Neelapu and Larry W. Kwak
    • Department of Lymphoma and Myeloma, Division of Cancer Medicine, The University of Texas, M.D. Anderson Cancer Center, Houston, Texas, USA.
Received December 07, 2006.

This is an Open Access article distributed under the terms of the Creative Commons Attribution Non-Commercial License (http://creativecommons.org/licenses/by-nc/3.0) which permits unrestricted noncommercial use, distribution, and reproduction in any medium, provided the original work is properly cited.

Abstract

The unique antigenic determinants (Idiotype [Id]) of the immunoglobulin expressed on a given B-cell malignancy can serve as a tumor-specific antigen for active immunotherapy. Therapeutic vaccines targeting the tumor-specific idiotype have demonstrated promising results against lymphomas in phase I/II studies and are currently being evaluated in phase III randomized trials. Additional vaccine therapies being developed include those based on DNA, dendritic cells, gene-modified tumor cells. It is hoped that immunotherapeutic agents, used in tandem or in combination, may in the future allow effective treatment of lymphoid malignancies and delay or even replace the need for conventional cytotoxic therapies.

Keywords
Idiotype; immunotherapy; lymphoma; vaccine

INTRODUCTION

Immunotherapy has now become an important part of our therapeutic armamentarium for hematologic malignancies. Several monoclonal antibodies have been approved by the FDA and are in widespread use either alone or in combination with chemotherapy or with other biologic agents. Other passive therapies with various immune cell populations are under investigation. Active immunotherapy, whereby the host is induced to make an immune response against its own tumor cells, has long been a goal of tumor immunologists.

The central hypothesis of active immunotherapy of cancer is that either the tumor cell itself or antigens derived from the tumor cell (which are specific, or at least selective, for the tumor cell) can be modified and injected back into the patient as a therapeutic (not preventive) vaccine. The desired result is activation of both major arms of the immune response, the host antibody response and potentially a host T-cell response, against the target tumor cell or antigen, thereby aiding in the eradication of the disease.

Idiotype as a tumor-specific antigen for B-cell Non-Hodgkin's lymphoma

Many of the efforts toward the development of a vaccine against human malignancies have been frustrated by the lack of identification of a tumor-specific antigen that would allow tumor cells to be distinguished from normal cells. In the case of B-cell NHL however, the tumor-specific immunoglobulin expressed on the surface of malignant B-cells can function as a tumor-specific antigen and has been exploited as a target for active immunotherapy. Each B lymphocyte expresses an immunoglobulin molecule on its surface, which is capable of recognizing and binding to a unique antigen. The variable region of the immunoglobulin that binds to the antigen is the product of a unique combination of gene sequences, and is referred to as its idiotype (Id) (Fig. 1). Since B-cell NHLs are composed of clonal proliferation of mature resting and reactive lymphocytes, which express synthesized immunoglobulins on the cell surface, the idiotypic determinants of the surface immunoglobulin of a B-cell lymphoma can serve as a tumor-specific marker for the malignant clone.

Fig. 1
Idiotype as a tumor antigen specific for B cell lymphoma. Malignancies of mature and resting B cells arise from clonal proliferation of cells that express immunoglobulins on their cell surface. Immunoglobulin molecules are composed of heavy and light chains, which possess highly specific variable regions at their amino termini and constant regions at their carboxy termini. The variable regions contain unique determinants termed idiotype (Id) that can be recognized as an antigen. The unique antigenic determinants are most likely derived from the hypervariable complementarity-determining regions (CDR), but not framework (FW) regions of the variable regions of heavy and light chains.18 Amino acid numbers in the variable heavy chain region are shown from the amino (NH2) terminus end.

Although not entirely applicable to Id, the major limitation to using self-tumor antigens for cancer vaccine is that they are protected by self-tolerance mechanisms. The first step in cancer vaccine development is therefore to answer the question of whether it is possible to render an inherently weak tumor antigen immunogenic. This is largely a scientific question, one that should be answered in the affirmative before asking the medical question of whether cancer vaccines can induce meaningful clinical benefit. In the case of lymphoma Id vaccine development, it has taken 10 years to answer this scientific question in a definitive manner.

Preclinical studies of Id vaccine

In the early 1970s, Lynch and Eisen demonstrated in mice that active immunization with purified immunoglobulins (Id) from mineral-oil-induced plasmacytomas (MOPCs) induced Id-specific tumor resistance.1, 2 This phenomenon has been reproduced subsequently in a number of lymphoma, myeloma, and leukemia models.3-11 In 1987, Kaminski et al., demonstrated that optimal immunization required conjugation to a strongly immunogenic carrier protein, such as KLH.12 Subsequently, Kwak and colleagues demonstrated that the use of GM-CSF as an adjuvant facilitated the induction of tumor-specific CD8+ T cells and enhanced the efficacy of the vaccine in a murine 38C13 lymphoma model.13 GM-CSF likely acts by recruiting and promoting maturation of professional antigen-presenting cells such as dendritic cells, which may in turn activate pathways of antigen processing that allow exogenous proteins to be presented by class I molecules.14 Animal studies also demonstrated that idiotypic vaccination conferred protection against tumor challenge and could cause regression of established tumor. Taken together, these results provided the rationale for testing autologous tumor-derived idiotypic surface immunoglobulin as a therapeutic "vaccine" against human B-cell NHL.

Phase I clinical trial of Id-KLH vaccine

The first human study of Id vaccination was pioneered by Kwak et al., in patients with follicular lymphoma (Table 1), which was a pilot study designed to determine whether it was possible to immunize against the Id portion of the protein.15 Follicular lymphoma patients in minimal residual disease or complete remission after chemotherapy, were immunized with subcutaneous injections of autologous purified tumor-derived immunoglobulin, conjugated to KLH. Because no Id-specific immune responses were observed before the addition of an immunologic adjuvant to the first group of patients, patients subsequently (nine patients) received the entire series of immunizations with a standard emulsion adjuvant (Syntex adjuvant formulation 1 - SAF-1). In all, 41 patients were treated on this pilot study; and 41% demonstrated specific anti-Id antibody and 17% demonstrated cellular proliferative responses.15 Of the 20 patients with residual disease following chemotherapy, two patients had complete regression of the tumor in association with the development of a specific immune response. Thus, these results were important because they demonstrated that patients with lymphoma could be induced to make sustained Id-specific immune responses by active immunization with purified autologous tumor-derived surface Ig conjugated to the immunogenic carrier KLH. Furthermore, the induction of Id-specific immune responses was demonstrated in the setting of minimal tumor burden after conventional chemotherapy.

Table 1
Published Clinical Trials of Idiotype Vaccination in Lymphoma

Phase II clinical trial of Id-KLH vaccine

Based on the preclinical observation that the addition of GM-CSF as an adjuvant to the vaccine induced tumor-specific CD8+ T cells,13 Bendandi and colleagues conducted a Phase II clinical trial where 20 previously untreated follicular lymphoma patients were treated with autologous tumor-derived Id-KLH+GM-CSF vaccine following induction of clinical remission with chemotherapy16 (Table 1). This produced a homogeneous group of patients, all in first complete remission (CR), who were given vaccine treatment in the setting of minimal residual disease. The vaccine was injected subcutaneously in 5 monthly doses starting approximately 6 months after completing chemotherapy to allow time for immunological recovery. The vaccine was well tolerated with the main adverse effects being injection site reactions such as erythema, induration, and pruritus. There were no long-term adverse effects due to the vaccine.

Following vaccination, anti-KLH antibody and cellular responses were induced in all patients. Anti-idiotype antibody responses were induced in 15 out of 20 (75%) patients and Id-specific and/or tumor-specific CD4+ and CD8+ T-cell responses were observed in 19 out of 20 (95%) patients.16 Importantly, significant levels of HLA class I-restricted killing of autologous tumor targets were also demonstrated in the vast majority of patients, suggesting the induction of a cytotoxic CD8+ T-cell response. The specificity for this response was shown by the lack of killing of nonneoplastic, normal B cells from the same patients.16 Further characterization of anti-idiotype cellular immune responses demonstrated that the T cells specifically recognized multiple unique immunodominant epitopes within the hypervariable complementarity-determining regions (CDR), but not framework regions of immunoglobulin heavy chain.17 Monitoring of the patients for minimal residual disease showed that 8 out of 11 patients with PCR-positive t(14;18) chromosomal translocation breakpoints converted to PCR negativity in their blood immediately after completing vaccination and sustained their molecular remissions for a median of 18+ months (range: 8+ to 32+ months).17 Thus, these results provided the first convincing evidence of an antitumor effect of Id vaccination. Analysis of time to relapse also provided an independent indication of clinical benefit. With a median follow-up of 9.2 years, median disease-free survival (DFS) is 8 years, and the overall survival rate is 95%.18 While definitive statements cannot be made, because this was not a randomized trial, the DFS appears superior to that of a historical, ProMACE chemotherapy-treated control group (median DFS, about 2.2 years).19

In conclusion, this Phase II clinical trial demonstrated that tumor-specific CD8+ T cells, capable of killing autologous tumor cells, could be induced by Id-KLH vaccination in combination with GM-CSF.17 This study also established GM-CSF as an essential component of this vaccine, as an earlier study using the same immunogen, administered without GM-CSF, showed humoral but no CD8+ T-cell responses.15

Phase III clinical trials of Id-KLH vaccine

The encouraging immunological and clinical outcome of the Id-KLH + GM-CSF vaccine in the Phase II clinical trial in patients with follicular lymphoma led to the initiation of a randomized double blind placebo controlled multicenter Phase III clinical trial to definitively answer the question of clinical benefit induced by idiotype vaccination. This Phase III trial was initiated by the National Cancer Institute, National Institutes of Health, Bethesda, MD and now sponsored by Biovest International, Inc. This trial was designed similar to the Phase II trial16 where previously untreated advanced stage follicular lymphoma patients initially underwent an excisional lymph node biopsy and were treated into clinical remission with a PACE chemotherapy regimen. Patients who achieve a CR or CRu (complete response unconfirmed) are randomized in a 2:1 manner either to the specific vaccination arm of Id-KLH+GM-CSF or the non-specific vaccination arm of KLH+GM-CSF. The primary endpoint for this trial is to compare the disease-free survival between the two arms.20 The secondary objectives of this trial are 1) to determine the ability of Id vaccine to produce a molecular CR in patients in clinical CR but PCR evidence of minimal residual disease after standard chemotherapy; 2) to evaluate the impact of Id vaccine to generate humoral and cellular immunologic responses against autologous tumor; and 3) to compare the overall survival of patients randomized to receive either the autologous tumor-derived Id vaccination (Id-KLH + GM-CSF) or non-specific vaccination (KLH + GM-CSF).

The Phase III trial was amended in July, 2006 to include administration of CHOP-R chemotherapy for remission induction prior to vaccine administration. The rationale for this was based on the fact that rituximab-based combination chemotherapy regimens were recently shown to induce a survival benefit in patients with follicular lymphoma and due to the fact that the induction of anti-tumor T cell immunity by idiotype vaccine was not impaired by the administration of prior rituximab containing chemotherapy. In the amended version of the trial, 540 patients will be randomized in a 2:1 ratio of Id-KLH vaccine + GM-CSF to KLH-KLH control vaccine + GM-CSF. The calculated trial size is sufficient to allow approximately 80% power to detect a difference between disease-free survival curves with an initial hazard ratio of 1.0 for the first 8 months and then an intended hazard ratio of 2.0 thereafter. The entire population of patients, whether induced with PACE or CHOP-R, will be analyzed in a combined manner. Patients will be stratified according to the International Prognostic Index, number of chemotherapy cycles, and type of chemotherapy.

Two additional randomized Phase III trials are currently evaluating the clinical efficacy of Id-KLH vaccine. These trials differ primarily in terms of the induction therapy and the method of idiotype production. The Genitope-sponsored trial uses cyclophosphamide, vincristine, and prednisone (CVP) chemotherapy,21 and the Favrille-sponsored trial uses the single agent anti-CD20 monoclonal antibody rituximab.22 Moreover, while only CR and CRu patients are vaccinated on the Biovest study, both CR, CRu, and PR patients are vaccinated on the Genitope trial, and CR, CRu, PR and stable disease patients are vaccinated on the Favrille trial. As opposed to the hybridoma method in the Biovest study, the Genitope and Favrille trials use recombinant DNA technology for production of idiotype protein, but with different immunoglobulin backbone structure and the cellular systems used for Id protein expression. Of these, Id protein of Genitope trial contains a human IgG3 heavy chain backbone is produced by plasmid transfection of murine lymphoma cells, whereas that of Favrille trial has IgG1 backbone and produced in insect cells. The results of interim analysis from these trials are expected within the next two years and it would be interesting to see whether the idiotype vaccines can induce clinically meaningful benefit in patients with both minimal residual disease (Biovest study) as well as with low tumor burden (Genitope and Favrille studies).

Second generation vaccines

Given that the general question of whether it is possible to immunize against Id has been answered by the completed phase II trial, and that a randomized controlled phase III clinical trial has been opened to answer the question of clinical efficacy, a third major research objective is to streamline the production of these individualized vaccines to make this therapy more practical. Accordingly, alternative methods for formulating Id, an otherwise nonimmunogenic antigen, into an immunogenic vaccine are being tested in preclinical syngeneic murine lymphoma models.

Liposomal idiotype vaccines

A strategy of replacing KLH with a more uniform liposomal carrier, containing dimyxistoyl phosphatidyl choline (DMPC) lipid and recombinant human interleukin (IL)-2 was explored and revealed that this liposomal carrier reproducibly converted lymphoma Id into a tumor rejection antigen.23 Head to head comparisons against Id-KLH (as well as Id-KLH + GM-CSF), controlled for Id antigen dose, revealed equivalent to superior potency for the liposomal vaccine. On the basis of these results, a pilot clinical trial of this formulation was performed and the results demonstrated that liposomal delivery of Id was well tolerated and induced sustained tumor-specific CD4+ and CD8+ T-cell responses in lymphoma patients.24

DNA vaccines

One of the major drawbacks of Id protein vaccines is that the vaccine production is time consuming and laborious. The idiotype for these protein vaccines is generated by hybridoma tissue culture technology (Fig. 2). An alternative to idiotype protein vaccination is to use DNA vaccines. Any delivery system that does not require protein expression holds tremendous potential for the goal of streamlining vaccine production. Immunoglobulin variable genes specific for the B-cell malignancies can be readily cloned25, 26 and combined into single chain variable fragment (scFv) format, encoding a single polypeptide consisting solely of VH and VL genes linked together inframe by a short, 15 amino acid linker. Preliminary studies in mice and humans showed that the DNA vaccine is weakly immunogenic in most cases and needs to be used together with an adjuvant to render it immunogenic. King et al. demonstrated that fusion of the gene encoding fragment C of tetanus toxin to scFv markedly enhances the anti-idiotypic antibody response and induced protection against B-cell lymphoma in mice.27, 28 Biragyn and colleagues showed that the efficiency of DNA vaccination in vivo could be greatly increased by encoding a fusion protein consisting of idiotype (scFv) fused to a proinflammatory chemokine moiety that facilitates targeting of antigen-presenting cells for chemokine receptor-mediated binding, uptake, and processing of scFv antigen for subsequent presentation to CD4+ or CD8+ T cells, or both.29-31 Specifically, mice immunized by gene gun with plasmids encoding monocyte chemotactic protein 3 (MCP-3) or interferon inducible protein 10 (IP-10)-scFv fusions, but not scFv alone, induced protective antitumor immunity against a large tumor challenge (20 times the minimum lethal dose). Furthermore, the level of protection was equivalent or superior to that of the prototype Id-KLH protein vaccine.

Fig. 2
Schematic diagram showing the production of Id protein vaccine using hybridoma technology. Id vaccines are custom-made from each patient's own tumor cells by fusion to the immortal myeloma cells. The Id protein is then chemically linked to the foreign protein KLH, combined with an immune-adjuvant and injected under the skin. IgVH, immunoglobulin heavy chain variable region; CDR3, complementarity-determining region 3.

Cell based therapeutic vaccine approach

Dendritic cell vaccines

In the past several years, DCs have been identified as the most powerful professional APCs. Dendritic cells can take up, process, and present antigen in the context of co-stimulatory signals required for activation of both CD4+ and CD8+ T cells. In recent years, several strategies have been developed to exploit the antigen-presenting properties of DCs. Timmerman et al demonstrated in a murine lymphoma model that vaccination with Id-KLH-pulsed DCs induced superior tumor-protective immunity than did native Id-pulsed DCs.32 In a pilot study, Hsu and colleagues used autologous DCs pulsed ex vivo with tumor-specific idiotype protein as a vaccine in 4 follicular B-cell lymphoma patients.33 Subsequent clinical trial on 35 patients with follicular lymphoma treated with the same strategy showed a 22% overall clinical response.34 This study demonstrated the feasibility and safety of Id-pulsed DCs as a vaccination strategy in humans.

Tumor cell vaccines

Immunization with irradiated, GM-CSF-transduced tumor cells can elicit cell mediated immunity against tumor antigens released by dying tumor cells and thereby resist growth of nontransfected tumor cells. Once again, GM-CSF serves to activate local antigen-presenting cells to efficiently take up and present these antigens to T-cells. In phase I/II studies of this approach in melanoma, renal cell carcinoma, and lung cancer, occasional clinical responses have been seen.35-37 Levitsky et al showed that immunization of mice with lymphoma cells genetically engineered to produce GM-CSF, and to a lesser extent cells producing IL-4, eradicated pre-established systemic lymphoma.38 The therapeutic effect of the GM-CSF- or IL-4-transfected lymphoma cells required both CD4+ and CD8+ T cells. In addition, the T-cell responses were shown to be Id specific in these mice, suggesting that GM-CSF-transduced tumor cell-based vaccination can induce immune responses against a native tumor antigen.

Conclusions and future prospects

Idiotype vaccination appears to be safe and immunogenic in patients with non-Hodgkin's lymphoma. The immune response appears to be directed against the tumor but not autologous normal B cells suggesting that idiotype is a truly tumor-specific antigen. Both humoral and cellular immune responses were shown to be independently associated with clinical responses following idiotype vaccination in patients with follicular lymphoma. Single arm Phase I and II idiotype vaccine trials demonstrated improved progression free survival compared with historical controls in patients with follicular lymphoma. However, data from ongoing randomized Phase III trials are needed to definitively determine the clinical benefit of idiotype vaccination in non-Hodgkin's lymphoma. If successful, idiotype vaccines are most likely to be used as an adjuvant following standard treatment with combination chemotherapy. Additionally, the recent demonstration of induction of antitumor T-cell responses by idiotype vaccination following B-cell depletion induced by rituximab39 suggests that idiotype vaccines can be used after administration of rituximab or rituximab-based chemotherapy. The combination of rituximab with idiotype vaccine would provide for the first time a combination biologic regimen for the treatment of this lymphoma. Indeed, the use of passively administered anti-tumor monoclonal antibodies such as rituximab with vaccines is likely to be complementary. Compared with monoclonal antibodies, vaccines are likely to target different tumor antigens, can induce immunological memory, and can induce polyclonal humoral and cellular immune responses, thereby minimizing the emergence of immune escape variants. Given that the median age of follicular lymphoma patients at diagnosis is 60 years, the development of such nontoxic immunotherapeutic approaches is highly desirable.

With the increased use of rituximab for the treatment of follicular lymphoma and other B-cell non-Hodgkin's lymphomas, further improvement in the potency of the idiotype vaccines would require strategies to enhance the T-cell responses since rituximab depletes normal B cells and impairs the generation of antibody responses following vaccination. Although novel adjuvants such as toll-like receptor ligands may prove to be more potent than cytokine adjuvants,40-42 further improvement of the cancer vaccines would probably also require disruption of the immunoregulatory pathways that modulate the magnitude and duration of the immune response. Studies in animal models suggest that the T-cell immune responses against foreign or self-antigens are regulated by several immunoregulatory pathways and/or peripheral tolerance mechanisms.43-45 For example, CD4+CD25+ regulatory T cells (Tregs) have been shown to downregulate T-cell responses against foreign antigens as well as tumor antigens, most of which are self-antigens.46 Similarly, cytotoxic T lymphocyte-associated antigen (CTLA)-4, a molecule that is expressed on activated T cells and Tregs, was shown to downregulate T-cell responsiveness and prevent the initiation and/or limit the magnitude of autoreactive T-cell responses.47 A host of other mechanisms such as programmed cell death 1 (PD-1), B7-H1, B7-H4 were recently described to negatively regulate T-cell responses.45 These new insights have led several investigators to hypothesize that the potency of cancer vaccines can be further enhanced by concurrent suppression or blocking of peripheral tolerance mechanisms and/or suppressive immunoregulatory pathways. Thus, depletion of CD4+CD25+ Tregs or blockade of CTLA-4, PD-1 or B7-H1 led to improved tumor control in various murine models.48-51 Moreover, simultaneous disruption of two immunoregulatory mechanisms by depletion of Tregs and blockade of CTLA-4 resulted in improved tumor rejection as compared with either one alone when used in combination with a tumor cell-based vaccine in a B16 melanoma model.51 These preclinical observations led to the initiation of pilot clinical trials using combination immunotherapeutic strategies analogous to combination chemotherapy that has been effectively used for the curative treatment of certain cancers including lymphoma. Dannull and colleagues have recently demonstrated that vaccine-mediated antitumor immunity is significantly enhanced in renal cell cancer patients after depletion of regulatory T cells using denileukin diftitox52 (a recombinant IL-2 diphtheria toxin conjugate; also known as Ontak); an FDA-approved drug for the treatment of cutaneous T-cell lymphomas. In another study, administration of a single dose of Ontak in ovarian cancer patients depleted Tregs and was associated with enhanced endogenous immunity.53 Similarly, blockade of CTLA-4 in combination with peptide vaccination has resulted in enhanced cancer immunity and durable objective responses in patients with metastatic melanoma.54, 55 Taken together, these preclinical and early phase clinical results support the evaluation of combination immunotherapy strategies in future clinical trials with idiotype vaccination for B cell lymphoma to stimulate an antitumor T-cell response and the simultaneous suppression of immune regulatory pathways to augment the induced T-cell response. The existence of multiple immune regulatory pathways necessitates systematic evaluation of these approaches in clinical trials to determine the optimal combination immunotherapy regimen.

ACKNOWLEDGEMENTS

The authors thank Allison F. Woo for review of manuscript.

References

    1. Lynch RG, Graff RJ, Sirisinha S, Simms ES, Eisen HN. Myeloma proteins as tumor-specific transplantation antigens. Proc Natl Acad Sci U S A 1972;69:1540–1544.
    1. Stevenson GT, Stevenson FK. Antibody to a molecularly-defined antigen confined to a tumour cell surface. Nature 1975;254:714–716.
    1. Sugai S, Palmer DW, Talal N, Witz IP. Protective and cellular immune responses to idiotypic determinants on cells from a spontaneous lymphoma of NZB/NZW F1 mice. J Exp Med 1974;140:1547–1558.
    1. Freedman PM, Autry JR, Tokuda S, Williams RC Jr. Tumor immunity induced by preimmunization with BALB/c mouse myeloma protein. J Natl Cancer Inst 1976;56:735–740.
    1. Stevenson GT, Elliott EV, Stevenson FK. Idiotypic determinants on the surface immunoglobulin of neoplastic lymphocytes: a therapeutic target. Fed Proc 1977;36:2268–2271.
    1. Bridges SH. Participation of the humoral immune system in the myeloma-specific transplantation resistance. J Immunol 1978;121:479–483.
    1. Daley MJ, Gebel HM, Lynch RG. Idiotype-specific transplantation resistance to MOPC-315: abrogation by postimmunization thymectomy. J Immunol 1978;120:1620–1624.
    1. Jorgensen T, Gaudernack G, Hannestad K. Immunization with the light chain and the VL domain of the isologous myeloma protein 315 inhibits growth of mouse plasmacytoma MOPC-315. Scand J Immunol 1980;11:29–35.
    1. Stevenson FK, Gordon J. Immunization with idiotypic immunoglobulin protects against development of B lymphocytic leukemia, but emerging tumor cells can evade antibody attack by modulation. J Immunol 1983;130:970–973.
    1. George AJ, Tutt AL, Stevenson FK. Anti-idiotypic mechanisms involved in the suppression of a mouse B cell lymphoma, BCL1. J Immunol 1987;138:628–634.
    1. Kwak LW, Campbell MJ, Zelenetz AD, Levy R. Combined syngeneic bone marrow transplantation and immunotherapy of a murine B-cell lymphoma: active immunization with tumor-derived idiotypic immunoglobulin. Blood 1990;76:2411–2417.
    1. Kaminski MS, Kitamura K, Maloney DG, Levy R. Idiotype vaccination against murine B cell lymphoma. Inhibition of tumor immunity by free idiotype protein. J Immunol 1987;138:1289–1296.
    1. Kwak LW, Young HA, Pennington RW, Weeks SD. Vaccination with syngeneic, lymphoma-derived immunoglobulin idiotype combined with granulocyte/macrophage colony-stimulating factor primes mice for a protective T-cell response. Proc Natl Acad Sci U S A 1996;93:10972–10977.
    1. Eager R, Nemunaitis J. GM-CSF gene-transduced tumor vaccines. Mol Ther 2005;12:18–27.
    1. Kwak LW, Campbell MJ, Czerwinski DK, Hart S, Miller RA, Levy R. Induction of immune responses in patients with B-cell lymphoma against the surface-immunoglobulin idiotype expressed by their tumors. N Engl J Med 1992;327:1209–1215.
    1. Bendandi M, Gocke CD, Kobrin CB, Benko FA, Sternas LA, Pennington R, et al. Complete molecular remissions induced by patient-specific vaccination plus granulocyte-monocyte colony-stimulating factor against lymphoma. Nat Med 1999;5:1171–1177.
    1. Baskar S, Kobrin CB, Kwak LW. Autologous lymphoma vaccines induce human T cell responses against multiple, unique epitopes. J Clin Invest 2004;113:1498–1510.
    1. Santos C, Stern L, Katz L, Watson T, Barry G. BiovaxID™ vaccine therapy of follicular lymphoma in first remission: long-term follow-up of a phase II trial and status of a controlled, randomized phase III trial. Blood 2005;106:2441.
      [abstract].
    1. Longo DL, Duffey PL, Gribben JG, Jaffe ES, Curti BD, Gause BL, et al. Combination chemotherapy followed by an immunotoxin (anti-B4-blocked ricin) in patients with indolent lymphoma: results of a phase II study. Cancer J 2000;6:146–150.
    1. Neelapu SS, Gause BL, Nikcevich DA, Schuster SJ, Winter J, Gockerman JP, et al. Phase III randomized trial of patient-specific vaccination for previously untreated patients with follicular lymphoma in first complete remission: Protocol summary and interim report. Clin Lymphoma 2005;6:61–64.
    1. Vose JM. Personalized immunotherapy for the treatment of non-Hodgkin's lymphoma: a promising approach. Hematol Oncol 2006;24:47–55.
    1. Hurvitz SA, Timmerman JM. Recombinant, tumour-derived idiotype vaccination for indolent B cell non-Hodgkin's lymphomas: a focus on FavId. Expert Opin Biol Ther 2005;5:841–852.
    1. Kwak LW, Pennington R, Boni L, Ochoa AC, Robb RJ, Popescu MC. Liposomal formulation of a self lymphoma antigen induces potent protective antitumor immunity. J Immunol 1998;160:3637–3641.
    1. Neelapu SS, Baskar S, Gause BL, Kobrin CB, Watson TM, Frye AR, et al. Human autologous tumor-specific T-cell responses induced by liposomal delivery of a lymphoma antigen. Clin Cancer Res 2004;10:8309–8317.
    1. Hawkins RE, Winter G, Hamblin TJ, Stevenson FK, Russell SJ. A genetic approach to idiotypic vaccination. J Immunother Emphasis Tumor Immunol 1993;14:273–278.
    1. Hawkins RE, Zhu D, Ovecka M, Winter G, Hamblin TJ, Long A, et al. Idiotypic vaccination against human B-cell lymphoma. Rescue of variable region gene sequences from biopsy material for assembly as single-chain Fv personal vaccines. Blood 1994;83:3279–3288.
    1. Spellerberg MB, Zhu D, Thompsett A, King CA, Hamblin TJ, Stevenson FK. DNA vaccines against lymphoma: promotion of anti-idiotypic antibody responses induced by single chain Fv genes by fusion to tetanus toxin fragment C. J Immunol 1997;159:1885–1892.
    1. King CA, Spellerberg MB, Zhu D, Rice J, Sahota SS, Thompsett AR, et al. DNA vaccines with single-chain Fv fused to fragment C of tetanus toxin induce protective immunity against lymphoma and myeloma. Nat Med 1998;4:1281–1286.
    1. Biragyn A, Tani K, Grimm MC, Weeks S, Kwak LW. Genetic fusion of chemokines to a self tumor antigen induces protective, T-cell dependent anti-tumor immunity. Nat Biotechnol 1999;17:253–258.
    1. Biragyn A, Surenhu M, Yang D, Ruffini RA, Haines BA, Klyushnenkova E, et al. Mediators of innate immunity that target immature, but not mature, dendritic cells induce antitumor immunity when genetically fused with nonimmunogenic tumor antigens. J Immunol 2001;167:6644–6653.
    1. Biragyn A, Ruffini PA, Coscia M, Harvey LK, Neelapu SS, Baskar S, et al. Chemokine receptor-mediated delivery directs self-tumor antigen efficiently into the class II processing pathway in vitro and induces protective immunity in vivo. Blood 2004;104:1961–1969.
    1. Timmerman JM, Levy R. Linkage of foreign carrier protein to a self-tumor antigen enhances the immunogenicity of a pulsed dendritic cell vaccine. J Immunol 2000;164:4797–4803.
    1. Hsu FJ, Benike C, Fagnoni F, Liles TM, Czerwinski D, Taidi B, et al. Vaccination of patients with B-cell lymphoma using autologous antigen-pulsed dendritic cells. Nat Med 1996;2:52–58.
    1. Timmerman JM, Czerwinski DK, Davis TA, Hsu FJ, Benike C, Hao ZM, et al. Idiotype-pulsed dendritic cell vaccination for B-cell lymphoma: clinical and immune responses in 35 patients. Blood 2002;99:1517–1526.
    1. Soiffer R, Lynch T, Mihm M, Jung K, Rhuda C, Schmollinger JC, et al. Vaccination with irradiated autologous melanoma cells engineered to secrete human granulocyte-macrophage colony-stimulating factor generates potent antitumor immunity in patients with metastatic melanoma. Proc Natl Acad Sci U S A 1998;95:13141–13146.
    1. Simons JW, Jaffee EM, Weber CE, Levitsky HI, Nelson WG, Carducci MA, et al. Bioactivity of autologous irradiated renal cell carcinoma vaccines generated by ex vivo granulocyte-macrophage colony-stimulating factor gene transfer. Cancer Res 1997;57:1537–1546.
    1. Nemunaitis J, Sterman D, Jablons D, et al. A phase I/II study of autologous GM-CSF gene-modified cancer vaccines in subjects with non-small cell lung cancer [abstract]. Proc Am Soc Clin Oncol 2001;20:254a.
      Abstract 1019.
    1. Levitsky HI, Montgomery J, Ahmadzadeh M, Staveley-O'Carroll K, Guarnieri F, Longo DL, et al. Immunization with granulocyte-macrophage colony-stimulating factor-transduced, but not B7-1-transduced, lymphoma cells primes idiotype-specific T cells and generates potent systemic antitumor immunity. J Immunol 1996;156:3858–3865.
    1. Neelapu SS, Kwak LW, Kobrin CB, Reynolds CW, Janik JE, Dunleavy K, et al. Vaccine-induced tumor-specific immunity despite severe B-cell depletion in mantle cell lymphoma. Nat Med 2005;11:986–991.
    1. Pashine A, Valiante NM, Ulmer JB. Targeting the innate immune response with improved vaccine adjuvants. Nat Med 2005;11 4 Suppl:S63–S68.
    1. Shackleton M, Davis ID, Hopkins W, Jackson H, Dimopoulos N, Tai T, et al. The impact of imiquimod, a Toll-like receptor-7 ligand (TLR7L), on the immunogenicity of melanoma peptide vaccination with adjuvant Flt3 ligand. Cancer Immun 2004;4:9.
    1. Speiser DE, Lienard D, Rufer N, Rubio-Godoy V, Rimoldi D, Lejeune F, et al. Rapid and strong human CD8+ T cell responses to vaccination with peptide, IFA, and CpG oligodeoxynucleotide 7909. J Clin Invest 2005;115:739–746.
    1. Walker LS, Abbas AK. The enemy within: keeping self-reactive T cells at bay in the periphery. Nat Rev Immunol 2002;2:11–19.
    1. Chen L. Co-inhibitory molecules of the B7-CD28 family in the control of T-cell immunity. Nat Rev Immunol 2004;4:336–347.
    1. Zou W. Immunosuppressive networks in the tumour environment and their therapeutic relevance. Nat Rev Cancer 2005;5:263–274.
    1. Sakaguchi S. Naturally arising Foxp3-expressing CD25+CD4+ regulatory T cells in immunological tolerance to self and non-self. Nat Immunol 2005;6:345–352.
    1. Carter LL, Carreno BM. Cytotoxic T-lymphocyte antigen-4 and programmed death-1 function as negative regulators of lymphocyte activation. Immunol Res 2003;28:49–59.
    1. Iwai Y, Ishida M, Tanaka Y, Okazaki T, Honjo T, Minato N. Involvement of PD-L1 on tumor cells in the escape from host immune system and tumor immunotherapy by PD-L1 blockade. Proc Natl Acad Sci U S A 2002;99:12293–12297.
    1. Iwai Y, Terawaki S, Honjo T. PD-1 blockade inhibits hematogenous spread of poorly immunogenic tumor cells by enhanced recruitment of effector T cells. Int Immunol 2005;17:133–144.
    1. Curiel TJ, Wei S, Dong H, Alvarez X, Cheng P, Mottram P, et al. Blockade of B7-H1 improves myeloid dendritic cell-mediated antitumor immunity. Nat Med 2003;9:562–567.
    1. Sutmuller RP, van Duivenvoorde LM, van Elsas A, Schumacher TN, Wildenberg ME, Allison JP, et al. Synergism of cytotoxic T lymphocyte-associated antigen 4 blockade and depletion of CD25(+) regulatory T cells in antitumor therapy reveals alternative pathways for suppression of autoreactive cytotoxic T lymphocyte responses. J Exp Med 2001;194:823–832.
    1. Dannull J, Su Z, Rizzieri D, Yang BK, Coleman D, Yancey D, et al. Enhancement of vaccine-mediated antitumor immunity in cancer patients after depletion of regulatory T cells. J Clin Invest 2005;115:3623–3633.
    1. Barnett B, Kryczek I, Cheng P, Zou W, Curiel TJ. Regulatory T cells in ovarian cancer: biology and therapeutic potential. Am J Reprod Immunol 2005;54:369–377.
    1. Phan GQ, Yang JC, Sherry RM, Hwu P, Topalian SL, Schwartzentruber DJ, et al. Cancer regression and autoimmunity induced by cytotoxic T lymphocyte-associated antigen 4 blockade in patients with metastatic melanoma. Proc Natl Acad Sci U S A 2003;100:8372–8377.
    1. Hodi FS, Mihm MC, Soiffer RJ, Haluska FG, Butler M, Seiden MV, et al. Biologic activity of cytotoxic T lymphocyte-associated antigen 4 antibody blockade in previously vaccinated metastatic melanoma and ovarian carcinoma patients. Proc Natl Acad Sci U S A 2003;100:4712–4717.
    1. Timmerman JM, Singh G, Hermanson G, Hobart P, Czerwinski DK, Taidi B, et al. Immunogenicity of a plasmid DNA vaccine encoding chimeric idiotype in patients with B-cell lymphoma. Cancer Res 2002;62:5845–5852.
    1. Barrios Y, Cabrera R, Yanez R, Briz M, Plaza A, Fores R, et al. Anti-idiotypic vaccination in the treatment of low-grade B-cell lymphoma. Haematologica 2002;87:400–407.
    1. Inoges S, Rodriguez-Calvillo M, Zabalegui N, Lopez-Diaz de Cerio A, Villanueva H, Soria E, et al. Clinical benefit associated with idiotypic vaccination in patients with follicular lymphoma. J Natl Cancer Inst 2006;98:1292–1301.
    1. Bertinetti C, Zirlik K, Heining-Mikesch K, Ihorst G, Dierbach H, Waller CF, et al. Phase I trial of a novel intradermal idiotype vaccine in patients with advanced B-cell lymphoma: specific immune responses despite profound immunosuppression. Cancer Res 2006;66:4496–4502.
    1. Redfern CH, Guthrie TH, Bessudo A, Densmore JJ, Holman PR, Janakiraman N, et al. Phase II trial of idiotype vaccination in previously treated patients with indolent non-Hodgkin's lymphoma resulting in durable clinical responses. J Clin Oncol 2006;24:3107–3102.

Metrics
Share
Figures

1 / 2

Tables

1 / 1

PERMALINK