Abstract
Recent insights into anti-tumor immunotherapy have led to a wave of clinical trials involving immunotherapy for lung cancer. Vaccines have evolved from nonspecific immune stimulants, like Bacillus Calmette-Guerin (BCG), to much more specific and potent strategies, some of which generate active immune responses against tumor-associated antigens. Understanding the mechanisms of anti-tumor immunity and identifying target antigens will likely improve these therapeutic strategies and provide them with a niche in the future of lung cancer therapy.
Keywords: Immunotherapy, Non-small cell lung cancer, Small cell lung cancer, Tumor immunology, Tumor vaccines
Lung cancer is the leading cause of cancer mortality, resulting in more than 160,000 deaths per year in the United States.1 Despite aggressive treatment with surgery, radiation and chemotherapy, the long-term survival for lung cancer patients remains low. Even those with “early stage” disease frequently succumb to lung cancer due to development of metastases. As a result, systemic therapy has become a requisite component of lung cancer management.
So far, systemic therapy has been limited to chemotherapy and biologic response modifiers. While novel agents like docetaxel, pemetrexed and erlotinib have recently demonstrated efficacy in treating patients with advanced lung cancer, clinical responses to treatment and improved survival have been modest. The limited successes of systemic chemotherapy have underscored the need to develop new therapeutic strategies like anti-tumor immunotherapy.
For several decades, effective immune-based anti-tumor therapy has been sought. There has been tantalizing evidence that the immune system, when properly stimulated, can eradicate cancer cells. Modest successes have been realized with nonspecific immune stimulants, such as interferon (IFN)-α and interleukin (IL)-2 for melanoma2 and renal cell carcinoma,3 and impressive anti-tumor activity has been observed with Bacille Calmette-Guerin (BCG) for non-invasive bladder cancer.4
Building on enthusiasm generated by nonspecific immunotherapy, antigen-specific anti-tumor agents have been tested in animal models and in humans with advanced carcinomas. These novel strategies have been shown to generate anti-tumor immune responses in prostate cancer,5 melanoma6–8 and non-Hodgkin’s lymphoma,9 although clinical benefit has rarely been observed. For lung cancer, immunotherapy has only recently advanced from nonspecific to antigen-specific vaccination.
Early Studies of Immunotherapy for Lung Cancer
Lung cancer immunotherapy before 1990 focused on nonspecific immune stimulants including BCG, thymosin10 and Corynebacterium parvum.11 Small cell lung cancer (SCLC) was commonly investigated because it was feasible to treat patients with minimal residual disease following curative chemotherapy and radiation.
Einhorn et al.12 reported a series of 58 SCLC patients treated during the 1970s with chemotherapy, radiation and BCG immunotherapy. Five of 19 patients (26%) with limited stage disease were long-term survivors. Two larger series of patients with lung cancer treated with BCG found improvements in survival compared to historical controls, as well as a survival benefit for intrapleural BCG injection for patients with pleural effusions.13,14 Patients with adenocarcinomas were reported to respond better than those with squamous cell carcinomas.15 Excitement about nonspecific agents was tempered, however, by a randomized study of BCG as adjuvant therapy for SCLC demonstrating no impact on overall survival.16 In addition, a study of preoperative intratumoral injection of BCG found no impact on disease-free or overall survival,17 however, both of these randomized trials were underpowered to detect small differences. The role of BCG in lung cancer immunotherapy is now primarily as an adjuvant immune stimulant with autologous tumor or antigen-specific vaccine strategies.18
T cell growth factor thymosin, another nonspecific immune stimulant, was found to have activity in combination with chemotherapy, especially among patients whose immune systems were not activated at the time therapy was initiated.10 Similarly, early studies using the bacterium Corynebacterium parvum as a systemic anti-tumor agent for non-small cell lung cancer (NSCLC) demonstrated dose-related tumor responses, but no effect on overall survival.11
During the 1990s nonspecific immunotherapy was attempted with IL-2 and other cytokines or inflammatory mediators. For example, Schiller et al.19 reported a series of 15 patients with advanced lung cancer treated with IL-2 and tumor necrosis factor-α. Cardiopulmonary toxicity was common, and there were no significant tumor responses. Similarly, a combination of IL-2 and IFN-α demonstrated no benefit.20 A combination of IL-2 and melatonin, on the other hand, demonstrated a clinical benefit (20% partial response, 50% stable disease) in a study of 20 patients.21
Targeting Lung Tumors with Passive Immunotherapy
Monoclonal antibodies, first produced more than 30 years ago, have only recently been applied to human anti-tumor therapy. “Humanized” antibodies targeting specific tumor-associated antigens are now widely embraced agents for therapy of lymphoma (e.g., rituximab) and breast cancer (e.g., trastuzumab). So far, however, passive immunotherapy has not made an impact on lung cancer.
Trastuzumab (anti-HER2 antibody) has been evaluated for patients with advanced NSCLC. In a phase II study of 24 patients with HER-2 overexpressing tumors, only one patient had a partial clinical response to therapy.22 In another study, trastuzumab was tested in combination with chemotherapy for advanced NSCLC.23 None of the 13 HER2-positive patients in this cohort responded to targeted therapy.
Another agent that has been developed recently is an antibody targeting the ganglioside fucosyl GM-1. In preclinical studies, this drug was shown to decrease formation of metastatic lung cancer via antibody-dependent cell-mediated cytotoxicity.24
Antigen-specific radioimmunotherapy has been attempted for SCLC by utilizing bispecific monoclonal antibodies. This was felt to be a promising treatment strategy because SCLC is highly radiosensitive. In one study, anti-carcinoembryonic antigen (CEA) antibody was attached to a radionucleotide-binding antibody; 3 of 12 patients responded to this treatment.25
Generating Active Anti-tumor Immunity Against Lung Cancer
While these “passive” immunotherapy strategies may lead to development of highly effective anti-tumor agents, the more potent therapeutic tools will likely be active immunotherapy agents. Lung cancer, although not very “immunogenic,” may provide an accessible target for the properly primed immune system.26
The identification of tumor-associated antigens has enabled development of vaccines that prime potent, antigen-specific immune responses. Such antigens were first identified for melanoma, initially by evaluating tumor-infiltrating lymphocytes. Melanoma has been considered an “immunogenic” tumor based on the presence of anti-tumor immune cells within tumor tissue, as well as the therapeutic benefits observed with nonspecific immune stimulatory agents like IL-2. Lung tumors have fewer tumor-infiltrating lymphocytes revealing that an effective immune response is not typically primed (not necessarily that it is primed and rendered ineffective).27 In lung cancer, as in melanoma, antigens targeted by tumor-infiltrating lymphocytes have been shown to induce tumor-specific cytolytic T cell responses.28,29 Identifying lung tumor antigens and presenting them in the optimal context may enable the immune system to generate anti-lung tumor effector cells that are usually absent.
Understanding how antigen-specific immune responses are generated has been a major focus of tumor immunology. As depicted in figure 1 ▶, antigen-presenting cells, including macrophages and dendritic cells, ingest parts of tumor cells, process their proteins and display them as short peptides bound to major histocompatibility complexes (MHC) class I and MHC class II. The T-cell receptor on naïve T cells binds to specific MHC/antigen complexes, leading to the exquisite specificity seen in cellular immunity. This T-cell receptor binding of the MHC/antigen complex, in conjunction with binding of costimulatory molecules B7.1 and B7.2 on the antigen-presenting cells, leads to activation of T cells. T cells that encounter their cognate antigen without proper costimulation (e.g., on a non-antigen-presenting cell) may be rendered tolerant, thus precluding generation of an immune response.
Figure 1.
Anti-tumor immune priming and response.
Helper T cells (CD4+) recognize their cognate antigens (MHC class II molecules) which are found only on antigen-presenting cells, whereas CD8+ cytolytic T cells recognize their cognate antigens (MHC class I molecules) that are found on all somatic cells including nonhematopoietic cells. Activation of CD4+ T cells leads to secretion of cytokines such as IFN-γ and IL-12, which in turn augment the stimulation of active CD8+ T cells. Helper T cells also augment the killing activity of natural killer cells, the phagocytic activity of macrophages and amplify antigen-specific immunity by local secretion of IL-2.
Priming anti-tumor immunity, therefore, requires knowledge of specific antigens, as well as delivery of appropriate immune signaling components, including costimulatory molecules and cytokines. Since some tumor-associated antigens have been identified for lung cancer, the field is now poised to develop potent anti-tumor vaccines that incorporate these other factors. Some that have been evaluated are discussed below.
Antigen-specific Vaccination
Now that tumor antigens like the ganglioside fucosyl GM-1 have been identified, targeted active immunotherapy strategies have become feasible (table 1 ▶). Krug et al.30 reported a clinical trial using a synthetic version of fucosyl GM-1 conjugated to the immune stimulant keyhole limpet hemocyanin in patients completing primary treatment for SCLC. Among 16 patients evaluated in this dose finding study, eight generated IgM responses. Another ganglioside, GD-3, has been tested in an active immunization strategy.31 BEC2-BCG, a vaccine that induces anti-ganglioside GD3 antibodies, was tested in SCLC patients after chemotherapy or combined chemotherapy and radiotherapy.16 In a large randomized international phase III trial, this agent provided no survival benefit.32
Table 1.
Human lung cancer vaccine trials.
Immunotherapeutic Agent | Investigator (ref) | Type of Trial |
Antigen-specific vaccines | ||
Fucosyl GM-1-KLH | Krug (30) | Phase I |
BEC2-BCG | Giaccone (18) | Phase III |
BLP25 (MUC1) | Palmer (35), Murray (36) | Phase II |
EGF (conjugated with P64K) | Gonzales (37), Neninger (38) | Phase II |
WT1 peptide | Oka (39) | Phase I |
MAGE-3 | Atanackovic (41) | Phase II |
P53/dendritic cells | Gabrilovich (44) | Phase II |
Strategies that incorporate cytokines or costimulatory molecules | ||
GVAX (GM-CSF) | Nemunaitis (52) | Phase I/II |
ALVAC-CEA/B7.1 | Ertl (61) | Phase II |
Allogeneic tumor cell with B7.1 | Raez (63) | Phase I |
Dendritic cell | Hirschowitz (62) | Phase I |
Polysialic acid, a long polymer of negatively charged sialic acid residues that binds to a neural cell adhesion molecule, has been found on the surface of SCLC tumor cells. Because this molecule has a role in embryonic development, immune tolerance is normally developed. To overcome this, polysialic acid was modified by N-propionylation. In a series of six patients vaccinated with the modified molecule, five demonstrated antibody responses against the target tumor antigen.33
Cancer-associated mucins are another potential target for immunotherapy. These molecules are thought to promote metastases by facilitating adhesion of malignant cells to the endothelial cell surface. They exhibit unique glycosylation patterns, making them tumor-specific immunogens.34 In a phase I clinical trial using MUC1 peptide in stage III/IV NSCLC,35 safety and tolerability of this agent was established. Five of 12 patients (42%) had immunologic responses, and 4 of 12 patients (33%) achieved stable disease. In a randomized phase II clinical trial including 65 patients with stage IIIB NSCLC, Murray et al.36 reported a 54% 2-year survival compared to a median survival of 13 months in the control group (best supportive care).
Epidermal growth factor (EGF), now a well-established target for biologic therapy, is also a potential tumor antigen. Preclinical studies have established the antigenicity and anti-tumor activity of EGF protein administered to animals.37 In two randomized phase II studies,38 recombinant EGF conjugated to Neisseria meningitides P64K protein was administered to a total of 40 advanced NSCLC patients. Anti-EGF antibody responses were identified with a significant increase in survival for patients who maintained antibody response (9.1 months vs. 4.5 months). The same agent was tested in a larger randomized phase II clinical trial38 which vaccinated 100 patients with stage IIIB or IV NSCLC who had progressed through first-line chemotherapy. Forty-five percent of vaccinated patients developed a strong anti-EGF antibody response. Compared to controls (best supportive care), those who received the treatment had significantly longer overall survival (8.5 vs. 4.3 months).
Wilm’s tumor protein (WT1), a protein overexpressed in leukemias and several solid tumors, including lung cancer, has been demonstrated to be a target antigen for immunotherapy. Oka et al.39 evaluated this vaccination strategy in 26 patients (10 with lung cancer). The patients received HLA-A*2402-restricted, natural or modified 9-mer WT1 peptide combined with Montanide ISA51 adjuvant. This study reported decreased tumor markers in 3 of 10 patients with lung cancer and identified a correlation between clinical response and anti-tumor CD8+ T cell activity. One responder continued to receive the treatment for 2 years without significant adverse effects.
MAGE-3, a tumor-associated antigen originally identified in melanoma, has also been found in non-small cell lung tumors.40 In a clinical trial,41 nine NSCLC patients were vaccinated with the protein; 3 developed antibody responses. Seven of 8 patients who received MAGE-3 combined with adjuvant ASO2B generated antibodies against MAGE-3. Several of these patients also developed T cell responses to the protein. The authors concluded that vaccination with this recombinant protein provides an integrated immune response against tumor cells.
Dendritic cells have recently been investigated as a delivery mechanism for tumor-associated antigens. Both whole cell42 and peptide43 strategies have been reported. In one study, 18 patients with metastatic gastrointestinal or lung cancer and HLA-A24 were treated with autologous dendritic cells pulsed with CEA-derived peptide. Immune reactions, measured by skin testing and in vitro T cell assays, were observed in most of the patients. Although no objective clinical responses were reported, some patients had stable disease while receiving this immunotherapy.
Another study of dendritic cell immunotherapy was reported by Gabrilovich et al.44 in which 22 patients with previously-treated extensive stage SCLC were treated with autologous dendritic cells transfected with adenovirus containing p53. Eleven of 20 patients (55%) demonstrated immunological response to vaccination. This study found few responders to the vaccine, but several patients responded to subsequent chemotherapy, suggesting a sensitizing effect.
We evaluated a novel strategy for priming immunity against tumor-antigens using heat shock proteins secreted by tumor cells.45 These proteins are “chaperones” that carry self- and tumor-specific peptides (potential tumor antigens). Initial studies with the heat shock protein gp96, isolated from human tumors, provided encouraging results in human trials.46,47 Our group has also investigated a gp96-Ig fusion protein, a chimeric protein that becomes secreted instead of remaining in the endoplasmic reticulum. Murine studies demonstrated that tumor-secreted gp96-Ig is capable of generating both immune and clinical responses. A phase I clinical trial of this agent is forthcoming.
Engineering More Potent Immunotherapy
The benefit of incorporating cytokines into anti-tumor vaccines has been well established. The cytokine granulocyte-monocyte colony stimulating factor (GM-CSF), a significant mediator of proliferation, maturation and migration of dendritic cells,48,49 has been shown to enhance the generation of potent, durable anti-tumor immunity.50 In a clinical trial using autologous, irradiated lung tumor (NSCLC) cells engineered to secrete GM-CSF,51 18 of 25 assessable patients demonstrated evidence of anti-tumor immunity. Three of 6 patients with metastases removed were found to have tumor necrosis and several patients in the study had long-term disease stability.
In a multicenter phase I/II trial, Nemunaitis et al.52 evaluated 43 lung cancer patients vaccinated with GVAX, a GM-CSF-secreting bystander cell admixed with autologous tumor lysate. The most common toxicity was a local injection-site reaction (93%). Three of 33 advanced-stage patients, 2 with bronchoalveolar carcinoma, had durable clinical responses. Longer median survival was observed in patients whose vaccines secreted more GM-CSF (17 months vs. 7 months), suggesting a cytokine dose-response relationship. However, measurements of immunologic response were not associated with clinical response or survival.
Another promising strategy is the incorporation of costimulatory molecules into tumor vaccines. Tumor cells transfected with B7.1 (figure 1 ▶) and HLA molecules have been shown to stimulate an immune response by direct antigen presentation and activation of T cells without intermediary cells.53 Vaccine trials with B7.1-transfected allogeneic or autologous cells have shown activity in murine and human studies of renal cell carcinoma,54–56 bladder cancer57 and breast cancer.58 In another clinical trial,59 an HLA-A21, HER2/neu-expressing allogeneic breast cancer cell line genetically modified to express B7.1 was administered as a vaccine to 30 women with previously-treated stage IV breast cancer. Immune responses were identified in 9 of 15 patients evaluated. Prolonged disease stabilization was observed in four patients, although no objective tumor regression was seen.
In a similar strategy, a non-replicating canarypox virus (ALVAC) was constructed to express both human CEA and the B7.1 costimulatory molecule.60 In a phase I trial, ALVAC-CEA-B7.1 was given without evidence of significant toxicity or autoimmune reactions. Three patients experienced clinically stable disease that correlated with increasing CEA-specific precursor T cells, as shown by in vitro IFN-γ enzyme-linked immunoassay spot tests. These three patients underwent repeated vaccinations resulting in augmented CEA-specific T cell responses. This strategy is now being tested in a phase II trial.61
In an attempt to incorporate more tumor-associated antigens, Hirschowitz et al.62 reported a novel strategy using dendritic cells pulsed with apoptotic bodies from an allogeneic NSCLC cell line expressing HER2/neu, CEA, WT1, MAGE2 and survivin. The cells were administered as an adjuvant agent to 16 patients with stage IA to IIIB NSCLC after surgery, chemoradiation or multimodality therapy. Six of the patients developed antigen-specific immune responses.
A more potent combination of target antigens may be delivered by an allogeneic whole cell-based vaccine. Such a vaccine could potentially target all the “shared” tumor-associated antigens and elicit a polyvalent cytolytic T lymphocyte response for each antigen, thus preventing tumor evasion by antigenic modulation. We developed an allogeneic lung cancer vaccine expressing the costimulatory molecule B7.163 to test the hypothesis that liberation of antigen in the context of the local inflammatory response will attract dendritic cells that uptake the antigen and the killed vaccine cells, and will cross-present the tumor-associated antigens through patient MHC to generate a tumor-specific cytolytic T lymphocyte response.
We treated 19 patients with irradiated allogeneic lung tumor (NSCLC) cells expressing the costimulatory molecule B7.1. Four patients experienced serious adverse events that were unrelated to vaccine. Another four patients experienced only minimal skin erythema. Among six clinically responding patients, immune responses were observed for as long as 150 weeks, long after administration of the vaccine. One patient had a partial clinical response and 5 had stable disease. Median survival for all patients was more than 18 months (90% confidence interval, 7 to 23 months), with estimated 1-year, 2-year and 3-year survivals of 52%, 30% and 30%, respectively. This study shows that a whole cell vaccine rendered immunogenic by transfection with B7 and HLA A1 or A2 can induce a potent immune response.
Challenges in Lung Cancer Immunotherapy
Despite the fact that many strategies have been evaluated, the most potent and effective lung cancer immunotherapy has not yet been identified because most agents that have shown preclinical efficacy have not been effective in human trials. The optimal source of antigens (e.g., autologous or allogeneic tumor-associated antigens), the role of costimulatory molecules like B7.1 and the mode of delivery (e.g., viral gene transfer, dendritic cells or tumor cells), are just some of the questions under investigation.
Another challenge with these agents is the lack of correlation between immune response and clinical activity. Clinical responses to immunotherapy may be delayed compared to clinical responses to chemotherapy. Also, agents that provoke an increase in tumor size (sometimes interpreted as tumor progression) may be provoking an inflammatory reaction within the tumors. Perhaps the classic ways that clinical responses are assessed (e.g., RECIST criteria) during chemotherapy may not be the most appropriate for immunotherapy trials. Instead, progression-free survival or overall survival may be better measures of clinical activity in these studies, which is something important to consider when designing or evaluating immunotherapy trials.
So far immunotherapy seems to hold the greatest promise in the adjuvant setting, when tumor burden is low and there is only minimal chance of significant immune tolerance. Another challenge, therefore, will be to identify the optimal sequence of adjuvant therapeutic agents. Adjuvant chemotherapy currently has an established, proven role as an adjuvant modality for early stage lung cancer, therefore, it will be important to study whether tumor vaccines will be as effective after chemotherapy.
Some new directions in anti-tumor immunotherapy will come from new studies of immune modulation. For example, a subset of CD4+ CD25+ T lymphocytes (known as regulatory T cells) have been shown to play a major role in down-regulating immune responses to tumors, including lung cancer.64,65 Depleting these cells with targeted monoclonal antibodies like denileukin diftitox may permit the generation of an anti-tumor immune response.66 Similarly, agents that target tolerizing B cells (e.g., rituximab) are being studied in the setting of anti-tumor immunotherapy.
Conclusion
Immunotherapy for lung cancer is a burgeoning therapeutic modality, although its development lags several years behind that for other malignancies. “Passive” immunity strategies like monoclonal antibodies have not yet demonstrated effectiveness for lung cancer. Targeted “active” immunotherapy, on the other hand, now includes a series of novel agents capable of generating anti-tumor immunity against lung cancer. Several agents have demonstrated immune responses, and some have been found to have clinical efficacy. In the future, improved treatments will likely come from identification of other tumor-associated antigens, as well as a more solid understanding of antigen presentation and costimulatory molecules. Targeted agents, in sequence with other adjuvant anti-tumor therapeutics, hold great promise for impacting the prognosis of lung cancer patients.
Grant Support: Career Development Award from the American Society of Clinical Oncology (ASCO) and RO1-CA39201-14
References
- 1.Jemal A, Murray T, Ward E, Samuels A, Tiwari RC, Ghafoor A, Feuer EJ, Thun MJ. Cancer statistics, 2005. CA Cancer J Clin 2005;55:10–30. [DOI] [PubMed] [Google Scholar]
- 2.Eton O, Legha SS, Bedikian AY, Lee JJ, Buzaid AC, Hodges C, Ring SE, Papadopoulos NE, Plager C, East MJ, Zhan F, Benjamin RS. Sequential biochemotherapy versus chemotherapy for metastatic melanoma: results from a phase III randomized trial. J Clin Oncol 2002;20:2045–2052. [DOI] [PubMed] [Google Scholar]
- 3.Coppin C, Porzsolt F, Awa A, Kumpf J, Coldman A, Wilt T. Immunotherapy for advanced renal cell cancer. Cochrane Database Syst Rev 2005;(1):CD001425. [DOI] [PubMed]
- 4.Alexandroff AB, Jackson AM, O’Donnell MA, James K. BCG immunotherapy of bladder cancer: 20 years on. Lancet 1999;353:1689–1694. [DOI] [PubMed] [Google Scholar]
- 5.Kaufman HL, Wang W, Manola J, DiPaola RS, Ko YJ, Sweeney C, Whiteside TL, Schlom J, Wilding G, Weiner LM. Phase II randomized study of vaccine treatment of advanced prostate cancer (E7897): a trial of the Eastern Cooperative Oncology Group. J Clin Oncol 2004;22:2122–2132. [DOI] [PubMed] [Google Scholar]
- 6.Zajac P, Oertli D, Marti W, Adamina M, Bolli M, Guller U, Noppen C, Padovan E, Schultz-Thater E, Heberer M, Spagnoli G. Phase I/II clinical trial of a nonreplicative vaccinia virus expressing multiple HLA-A0201-restricted tumor-associated epitopes and costimulatory molecules in metastatic melanoma patients. Hum Gene Ther 2003;14:1497–1510. [DOI] [PubMed] [Google Scholar]
- 7.Schadendorf D, Paschen A, Sun Y. Autologous, allogeneic tumor cells or genetically engineered cells as cancer vaccine against melanoma. Immunol Lett 2000;74:67–74. [DOI] [PubMed] [Google Scholar]
- 8.Quan WD Jr, Dean GE, Spears L, Spears CP, Groshen S, Merritt JA, Mitchell MS. Active specific immunotherapy of metastatic melanoma with an antiidiotype vaccine: a phase I/II trial of I-Mel-2 plus SAF-m. J Clin Oncol 1997;15:2103–2110. [DOI] [PubMed] [Google Scholar]
- 9.Kwak LW. Translational development of active immunotherapy for hematologic malignancies. Semin Oncol 2003;30(suppl):17–22. [DOI] [PubMed] [Google Scholar]
- 10.Lipson SD, Chretien PB, Makuch R, Kenady DE, Cohen MH. Thymosin immunotherapy in patients with small cell carcinoma of the lung: correlation of in vitro studies with clinical course. Cancer 1979;43:863–870. [DOI] [PubMed] [Google Scholar]
- 11.Issell BF, Valdivieso M, Hersh EM, Richman S, Gutterman JU, Bodey GP. Combination chemoimmunotherapy for extensive non-oat cell lung cancer. Cancer Treat Rep 1978;62:1059–1063. [PubMed] [Google Scholar]
- 12.Einhorn LH, Bond WH, Hornback N, Joe BT. Long-term results in combined-modality treatment of small cell carcinoma of the lung. Semin Oncol 1978;5:309–313. [PubMed] [Google Scholar]
- 13.Yasumoto K, Manabe H, Yanagawa E, Nagano N, Ueda H, Hirota N, Ohta M, Nomoto K, Azuma I, Yamamura Y. Nonspecific adjuvant immunotherapy of lung cancer with cell wall skeleton of Mycobacterium bovis Bacillus Calmette-Guerin. Cancer Res 1979;39:3262–3267. [PubMed] [Google Scholar]
- 14.Yamamura Y, Sakatani M, Ogura T, Azuma I. Adjuvant immunotherapy of lung cancer with BCG cell wall skeleton (BCG-CWS). Cancer 1979;43:1314–1319. [DOI] [PubMed] [Google Scholar]
- 15.Hadziev S, Mandulova P, Kavaklieva-Dimitrova J, Penev K, Spassova M, Madzarova S. BCG immunotherapy of lung cancer in a district oncology dispensary. I. Study of 860 patients with histologic diagnosis. Neoplasma 1982;29:93–110. [PubMed] [Google Scholar]
- 16.Maurer LH, Pajak T, Eaton W, Comis R, Chahinian P, Faulkner C, Silberfarb PM, Henderson E, Rege VB, Baldwin PE, Weiss R, Rafla S, Prager D, Carey R, Perry M, Choi NC. Combined modality therapy with radiotherapy, chemotherapy, and immunotherapy in limited small-cell carcinoma of the lung: a Phase III cancer and Leukemia Group B Study. J Clin Oncol 1985;3:969–976. [DOI] [PubMed] [Google Scholar]
- 17.Matthay RA, Mahler DA, Beck GJ, Loke J, Baue AE, Carter DC, Mitchell MS. Intratumoral Bacillus Calmette-Guerin immunotherapy prior to surgery for carcinoma of the lung: results of a prospective randomized trial. Cancer Res 1986;46:5963–5968. [PubMed] [Google Scholar]
- 18.Giaccone G, Debruyne C, Felip E, Millward M, D’Addario G, Thiberville L, Rome L, Zatloukal P, Legrand C; Phase III study of BEC2/BCG vaccination in limited disease small cell lung cancer (LD-SCLC) patients, following response to chemotherapy and thoracic irradiation (EORTC 08971, the SILVA study). J Clin Oncol 2004 ASCO Annual Meeting Proceedings (Post-Meeting Edition);22(14S):7020.
- 19.Schiller JH, Morgan-Ihrig C, Levitt ML. Concomitant administration of interleukin-2 plus tumor necrosis factor in advanced non-small cell lung cancer. Am J Clin Oncol 1995;18:47–51. [DOI] [PubMed] [Google Scholar]
- 20.Jansen RL, Slingerland R, Goey SH, Franks CR, Bolhuis RL, Stoter G. Interleukin-2 and interferon-alpha in the treatment of patients with advanced non-small-cell lung cancer. J Immunother 1992;12:70–73. [DOI] [PubMed] [Google Scholar]
- 21.Lissoni P, Tisi E, Barni S, Ardizzoia A, Rovelli F, Rescaldani R, Ballabio D, Benenti C, Angeli M, Tancini G, Conti A, Maestroni GJ. Biological and clinical results of a neuroimmunotherapy with interleukin-2 and the pineal hormone melatonin as a first line treatment in advanced non-small cell lung cancer. Br J Cancer 1992;66:155–158. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 22.Clamon G, Herndon J, Kern J, Govindan R, Garst J, Watson D, Green M; Cancer and Leukemia Group B. Lack of trastuzumab activity in nonsmall cell lung carcinoma with overexpression of erb-B2: 39810: a phase II trial of Cancer and Leukemia Group B. Cancer 2005;103:1670–1675. [DOI] [PubMed] [Google Scholar]
- 23.Lara PN Jr, Laptalo L, Longmate J, Lau DH, Gandour-Edwards R, Gumerlock PH, Doroshow JH, Gandara DR; California Cancer Consortium. Trastuzumab plus docetaxel in HER2/neu-positive non-small-cell lung cancer: a California Cancer Consortium screening and phase II trial. Clin Lung Cancer 2004;5:231–236. [DOI] [PubMed] [Google Scholar]
- 24.Hanibuchi M, Yano S, Nishioka Y, Yanagawa H, Kawano T, Sone S. Therapeutic efficacy of mouse-human chimeric anti-ganglioside GM2 monoclonal antibody against multiple organ micrometastases of human lung cancer in NK cell-depleted SCID mice. Int J Cancer 1998;78:480–485. [DOI] [PubMed] [Google Scholar]
- 25.Vuillez JP, Kraeber-Bodere F, Moro D, Bardies M, Douillard JY, Gautherot E, Rouvier E, Barbet J, Garban F, Moreau P, Chatal JF. Radioimmunotherapy of small cell lung carcinoma with the two-step method using a bispecific anti-carcinoembryonic antigen/anti-diethylenetriaminepentaacetic acid (DTPA) antibody and iodine-131 Di-DTPA hapten: results of a phase I/II trial. Clin Cancer Res 1999;5(suppl):3259S–3267S. [PubMed] [Google Scholar]
- 26.Liu B, Podack ER, Allison JP, Malek TR. Generation of primary tumor-specific CTL in vitro to immunogenic and poorly immunogenic mouse tumors. J Immunol 1996;156:1117–1125. [PubMed] [Google Scholar]
- 27.Wakabayashi O, Yamazaki K, Oizumi S, Hommura F, Kinoshita I, Ogura S, Dosaka-Akita H, Nishimura M. CD4+ T cells in cancer stroma, not CD8+ T cells in cancer cell nests, are associated with favorable prognosis in human non-small cell lung cancers. Cancer Sci 2003;94:1003–1009. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 28.Kawano K, Gomi S, Tanaka K, Tsuda N, Kamura T, Itoh K, Yamada A. Identification of a new endoplasmic reticulum-resident protein recognized by HLA-A24-restricted tumor-infiltrating lymphocytes of lung cancer. Cancer Res 2000;60:3550–3558. [PubMed] [Google Scholar]
- 29.Nishizaka S, Gomi S, Harada K, Oizumi K, Itoh K, Shichijo S. A new tumor-rejection antigen recognized by cytotoxic T lymphocytes infiltrating into a lung adenocarcinoma. Cancer Res 2000;60:4830–4837. [PubMed] [Google Scholar]
- 30.Krug LM, Ragupathi G, Hood C, Kris MG, Miller VA, Allen JR, Keding SJ, Danishefsky SJ, Gomez J, Tyson L, Pizzo B, Baez V, Livingston PO. Vaccination of patients with small-cell lung cancer with synthetic fucosyl GM-1 conjugated to keyhole limpet hemocyanin. Clin Cancer Res 2004;10:6094–6100. [DOI] [PubMed] [Google Scholar]
- 31.Chapman PB. Vaccinating against GD3 ganglioside using BEC2 anti-idiotypic monoclonal antibody. Curr Opin Investig Drugs 2003;4:710–715. [PubMed] [Google Scholar]
- 32.Grant SC, Kris MG, Houghton AN, Chapman PB. Long survival of patients with small cell lung cancer after adjuvant treatment with the anti-idiotypic antibody BEC2 plus Bacillus Calmette-Guerin Clin Cancer Res 1999;5:1319–1323. [PubMed] [Google Scholar]
- 33.Krug LM, Ragupathi G, Ng KK, Hood C, Jennings HJ, Guo Z, Kris MG, Miller V, Pizzo B, Tyson L, Baez V, Livingston PO. Vaccination of small cell lung cancer patients with polysialic acid or N-propionylated polysialic acid conjugated to keyhole limpet hemocyanin. Clin Cancer Res 2004;10:916–923. [DOI] [PubMed] [Google Scholar]
- 34.Ding L, Lalani EN, Reddish M, Koganty R, Wong T, Samuel J, Yacyshyn MB, Meikle A, Fung PY, Taylor-Papadimitriou J, Longenecker BM. Immunogenicity of synthetic peptides related to the core peptide sequence encoded by the human MUC1 mucin gene: effect of immunization on the growth of murine mammary adenocarcinoma cells transfected with the human MUC1 gene. Cancer Immunol Immunother 1993;36:9–17. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 35.Palmer M, Parker J, Modi S, Butts C, Smylie M, Meikle A, Kehoe M, MacLean G, Longenecker M. Phase I study of the BLP25 (MUC1 peptide) liposomal vaccine for active specific immunotherapy in stage IIIB/IV non-small-cell lung cancer. Clin Lung Cancer 2001;3:49–57. [DOI] [PubMed] [Google Scholar]
- 36.Murray N, Butts C, Maksymiuk A, Marshall E, Goss C, Soulieres D. L-BLP25 Non-Small Cell Lung Cancer Study Group A liposomal MUC1 vaccine for treatment of non-small cell lung cancer (NSCLC); updated survival results from patients with stage IIIB disease. Proc Am Soc Clin Oncol 2005;23 (Abstr 7037).
- 37.Gonzalez G, Crombet T, Torres F, Catala M, Alfonso L, Osorio M, Neninger E, Garcia B, Mulet A, Perez R, Lage R. Epidermal growth factor-based cancer vaccine for non-small-cell lung cancer therapy. Ann Oncol 2003;14:461–466. [DOI] [PubMed] [Google Scholar]
- 38.Neninger E, Crombet T, Osorio M, Catala M, Torre A, Leonard I, García B, Marinillo P, González G, Lage A. Vaccination with EGF active immunotherapy improves survival in advanced non small cell lung cancer (NSCLC) patients: interim analysis of a randomized Phase II trial. Proc Am Soc Clin Oncol 2005;23 (Abstr 7210).
- 39.Oka Y, Tsuboi A, Taguchi T, Osaki T, Kyo T, Nakajima H, Elisseeva OA, Oji Y, Kawakami M, Ikegame K, Hosen N, Yoshihara S, Wu F, Fujiki F, Murakami M, Masuda T, Nishida S, Shirakata T, Nakatsuka S, Sasaki A, Udaka K, Dohy H, Aozasa K, Noguchi S, Kawase I, Sugiyama H. Induction of WT1 (Wilms’ tumor gene)-specific cytotoxic T lymphocytes by WT1 peptide vaccine and the resultant cancer regression. Proc Natl Acad Sci U S A 2004;101:13885–13890. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 40.Sienel W, Varwerk C, Linder A, Kaiser D, Teschner M, Delire M, Stamatis G, Passlick B. Melanoma associated antigen (MAGE)-A3 expression in Stages I and II non-small cell lung cancer: results of a multi-center study. Eur J Cardiothorac Surg 2004;25:131–134. [DOI] [PubMed] [Google Scholar]
- 41.Atanackovic D, Altorki NK, Stockert E, Williamson B, Jungbluth AA, Ritter E, Santiago D, Ferrara CA, Matsuo M, Selvakumar A, Dupont B, Chen YT, Hoffman EW, Ritter G, Old LJ, Gnjatic S. Vaccine-induced CD4+ T cell responses to MAGE-3 protein in lung cancer patients. J Immunol 2004;172:3289–3296. [DOI] [PubMed] [Google Scholar]
- 42.Zhou Y, McEarchern JA, Howard E, Pestano G, Salgaller ML, Bosch ML. Dendritic cells efficiently acquire and present antigen derived from lung cancer cells and induce antigen-specific T-cell responses. Cancer Immunol Immunother 2003;52:413–422. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 43.Ueda Y, Itoh T, Nukaya I, Kawashima I, Okugawa K, Yano Y, Yamamoto Y, Naitoh K, Shimizu K, Imura K, Fuji N, Fujiwara H, Ochiai T, Itoi H, Sonoyama T, Hagiwara A, Takesako K, Yamagishi H. Dendritic cell-based immunotherapy of cancer with carcinoembryonic antigen-derived, HLA-A24-restricted CTL epitope: Clinical outcomes of 18 patients with metastatic gastrointestinal or lung adenocarcinomas. Int J Oncol 2004;24:909–917. [PubMed] [Google Scholar]
- 44.Gabrilovich DI, Mirza N, Chiappori A, Dunn M, Willis M, Janssen W, Smilee R, Menander K, Chada S, Antonia S. Initial results of a Phase II trial of patients with extensive stage small cell lung cancer (SCLC) immunized with dendritic cells (DC) transduced with wild-type p53. Proc Am Soc Clin Oncol 2005;23 (Abstr 2543).
- 45.Yamazaki K, Nguyen T, Podack ER. Cutting edge: tumor secreted heat shock-fusion protein elicits CD8 cells for rejection. J Immunol 1999;163:5178–5182. [PubMed] [Google Scholar]
- 46.Janetzki S, Palla D, Rosenhauer V, Lochs H, Lewis JJ, Srivastava PK. Immunization of cancer patients with autologous cancer-derived heat shock protein gp96 preparations: a pilot study. Int J Cancer 2000;88:232–238. [DOI] [PubMed] [Google Scholar]
- 47.Anderson KM, Srivastava PK. Heat, heat shock, heat shock proteins and death: a central link in innate and adaptive immune responses. Immunol Lett 2000;74:35–39. [DOI] [PubMed] [Google Scholar]
- 48.Beyer J, Schwella N, Zingsem J, Strohscheer I, Schwaner I, Oettle H, Serke S, Huhn D, Stieger W. Hematopoietic rescue after high-dose chemotherapy using autologous peripheral-blood progenitor cells or bone marrow: a randomized comparison. J Clin Oncol 1995;13:1328–1335. [DOI] [PubMed] [Google Scholar]
- 49.Stone RM, Berg DT, George SL, Dodge RK, Paciucci PA, Schulman P, Lee EJ, Moore JO, Powell BL, Schiffer CA. Granulocyte-macrophage colony-stimulating factor after initial chemotherapy for elderly patients with primary acute myelogenous leukemia. Cancer and Leukemia Group B. N Engl J Med 1995;332:1671–1677. [DOI] [PubMed] [Google Scholar]
- 50.Dranoff G, Jaffee E, Lazenby A, Golumbek P, Levitsky H, Brose K, Jackson V, Hamada H, Pardoll D, Mulligan RC. Vaccination with irradiated tumor cells engineered to secrete murine granulocyte-macrophage colony-stimulating factor stimulates potent, specific, and long-lasting anti-tumor immunity. Proc Natl Acad Sci U S A 1993;90:3539–3543. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 51.Salgia R, Lynch T, Skarin A, Lucca J, Lynch C, Jung K, Hodi FS, Jaklitsch M, Mentzer S, Swanson S, Lukanich J, Bueno R, Wain J, Mathisen D, Wright C, Fidias P, Donahue D, Clift S, Hardy S, Neuberg D, Mulligan R, Webb I, Sugarbaker D, Mihm M, Dranoff G. Vaccination with irradiated autologous tumor cells engineered to secrete granulocyte-macrophage colony-stimulating factor augments antitumor immunity in some patients with metastatic non-small-cell lung carcinoma. J Clin Oncol 2003;21:624–630. [DOI] [PubMed] [Google Scholar]
- 52.Nemunaitis J, Sterman D, Jablons D, Smith JW 2nd, Fox B, Maples P, Hamilton S, Borellini F, Lin A, Morali S, Hege K. Granulocyte-macrophage colony-stimulating factor gene-modified autologous tumor vaccines in non-small-cell lung cancer. J Natl Cancer Inst 2004;96:326–331. [DOI] [PubMed] [Google Scholar]
- 53.Johnston JV, Malacko AR, Mizuno MT, McGowan P, Hellstrom I, Hellstrom KE, Marquardt H, Chen L. B7-CD28 costimulation unveils the hierarchy of tumor epitopes recognized by major histocompatibility complex class I-restricted CD8+ cytolytic T lymphocytes. J Exp Med 1996;183:791–800. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 54.Antonia SJ, Seigne J, Diaz J, Muro-Cacho C, Extermann M, Farmelo MJ, Friberg M, Alsarraj M, Mahany JJ, Pow-Sang J, Cantor A, Janssen W. Phase I trial of a B7-1 (CD80) gene modified autologous tumor cell vaccine in combination with systemic interleukin-2 in patients with metastatic renal cell carcinoma. J Urol 2002;167:1995–2000. [PubMed] [Google Scholar]
- 55.Antonia SJ, Seigne JD. B7-1 gene-modified autologous tumor-cell vaccines for renal-cell carcinoma. World J Urol 2000;18:157–163. [DOI] [PubMed] [Google Scholar]
- 56.Wang J, Nakamoto T, Kasaoka Y, Usui T, Hamada H. Anti-tumor effect of murine renal cell carcinoma cells genetically modified to express B7-1 combined with cytokine secreting fibroblasts. Hiroshima J Med Sci 2000;49:73–82. [PubMed] [Google Scholar]
- 57.Loskog A, Bjorkland A, Brown MP, Korsgren O, Malmstrom PU, Totterman TH. Potent antitumor effects of CD154 transduced tumor cells in experimental bladder cancer. J Urol 2001;166:1093–1097. [PubMed] [Google Scholar]
- 58.Wu RS, Kobie JJ, Besselsen DG, Fong TC, Mack VD, McEarchern JA, Akporiaye ET. Comparative analysis of IFN-gamma B7.1 and antisense TGF-beta gene transfer on the tumorigenicity of a poorly immunogenic metastatic mammary carcinoma. Cancer Immunol Immunother 2001;50:229–240. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 59.Dols A, Smith JW 2nd, Meijer SL, Fox BA, Hu HM, Walker E, Rosenheim S, Moudgil T, Doran T, Wood W, Seligman M, Alvord WG, Schoof D, Urba WJ. Vaccination of women with metastatic breast cancer, using a costimulatory gene (CD80)-modified, HLA-A2-matched, allogeneic, breast cancer cell line: clinical and immunological results. Hum Gene Ther 2003;14:1117–1123. [DOI] [PubMed] [Google Scholar]
- 60.Horig H, Lee DS, Conkright W, Divito J, Hasson H, LaMare M, Rivera A, Park D, Tine J, Guito K, Tsang KW, Schlom J, Kaufman HL. Phase I clinical trial of a recombinant canarypoxvirus (ALVAC) vaccine expressing human carcinoembryonic antigen and the B7.1 co-stimulatory molecule. Cancer Immunol Immunother 2000;49:504–514. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 61.Ertl HC. Technology evaluation: ALVAC-CEA/B7.1, Aventis Pasteur/Therion. Curr Opin Mol Ther 2002;4:601–605. [PubMed] [Google Scholar]
- 62.Hirschowitz EA, Foody T, Kryscio R, Dickson L, Sturgill J, Yannelli J. Autologous dendritic cell vaccines for non-small-cell lung cancer J Clin Oncol 2004;22:2808–2815. [DOI] [PubMed] [Google Scholar]
- 63.Raez LE, Cassileth PA, Schlesselman JJ, Sridhar K, Padmanabhan S, Fisher EZ, Baldie PA, Podack ER. Allogeneic vaccination with a B7.1 HLA-A gene-modified adenocarcinoma cell line in patients with advanced non-small-cell lung cancer. J Clin Oncol 2004;22:2800–2807. [DOI] [PubMed] [Google Scholar]
- 64.Onizuka S, Tawara I, Shimizu J, Sakaguchi S, Fujita T, Nakayama E. Tumor rejection by in vivo administration of anti-CD25 (interleukin-2 receptor alpha) monoclonal antibody. Cancer Res 1999;59:3128–3133. [PubMed] [Google Scholar]
- 65.Shimizu J, Yamazaki S, Sakaguchi S. Induction of tumor immunity by removing CD25+CD4+ T cells: a common basis between tumor immunity and autoimmunity. J Immunol 1999;163:5211–5218. [PubMed] [Google Scholar]
- 66.Vieweg J, Su Z, Dannull J. Enhancement of antitumor immunity following depletion of CD4+CD25+ regulatory T-cells. J Clin Oncol 2004, ASCO Annual Meeting Proceedings (Post-Meeting Edition);22(14S):2506.