3,010
Views
83
CrossRef citations to date
0
Altmetric
Review

Allogeneic tumor cell vaccines

The promise and limitations in clinical trials

, , , , , & show all
Pages 52-63 | Received 30 Apr 2013, Accepted 23 Sep 2013, Published online: 24 Sep 2013

Abstract

The high mortality rate associated with cancer and its resistance to conventional treatments such as radiation and chemotherapy has led to the investigation of a variety of anti-cancer immunotherapies. The development of novel immunotherapies has been bolstered by the discovery of tumor-associated antigens (TAAs), through gene sequencing and proteomics. One such immunotherapy employs established allogeneic human cancer cell lines to induce antitumor immunity in patients through TAA presentation. Allogeneic cancer immunotherapies are desirable in a clinical setting due to their ease of production and availability. This review aims to summarize clinical trials of allogeneic tumor immunotherapies in various cancer types. To date, clinical trials have shown limited success due potentially to extensive degrees of inter- and intra-tumoral heterogeneity found among cancer patients. However, these clinical results provide guidance for the rational design and creation of more effective allogeneic tumor immunotherapies for use as monotherapies or in combination with other therapies.

Introduction

Cancer is characterized by the uncontrolled proliferation of the body’s cells.Citation1,Citation2 Initiated to a great extent by mutations in the genome affecting cell regulatory function, potential cancers arise throughout the body constantly.Citation3 However, natural processes such as apoptosis and immune surveillance eradicate these cancers before the development of tumors.Citation4-Citation10 Despite the protection it confers, immune surveillance directs the selection of malignancies. This process, termed immunoediting, explains the mechanism by which cancers arise in immune competent individuals. Briefly, malignant cells recognized by the immune system are eliminated, while unrecognized malignancies survive. The repeated selection of these immunologically silent, cancerous cells leads to the eventual formation of a tumor mass. This tumor microenvironment is immunosuppressive due to the secretion of inhibitory cytokines, expression of inhibitory membrane bound ligands and recruitment of regulatory cell populations that deactivate tumor-specific immune cells.Citation5,Citation11,Citation12 These factors create an environment which promotes cancer growth, unhindered by the body’s natural defenses.Citation5

Although current cancer therapies such as chemotherapeutic agents and ionizing radiation decrease the percentage of relapse in some types of cancers, the clones that emerge after treatment may be more resistant to the same treatments. Further, these therapies are also associated with either a lack of specificity in the case of chemotherapy or a loss of hematopoietic potential following radiation therapy.Citation13 While these problems remain unsolved, activation of the immune system against cancer remains as a viable therapeutic option.Citation14-Citation17 As a result, a number of immunotherapeutic treatments have thus been explored to augment a patient’s natural anti-tumor immune response and overcome tumor-induced immunosuppression. These therapies, varying greatly in composition, aim to induce specific anti-tumor immunity.

Allogeneic vs. Autologous Therapeutic Cancer Vaccines

Therapeutic vaccination strategies for cancer can be categorized broadly by antigen source. From an immunological standpoint, successful cancer therapeutics stimulate the immune system against a broad range of tumor-associated antigens (TAAs) while conferring lasting immunity. Other considerations include vaccine efficacy over multiple doses, vaccine kinetics, and specificity. The source of biological material for a vaccine can modulate these properties and influence the vaccine’s overall efficacy.

Autologous cancer vaccination strategies use material derived from a patient’s tumor to create personalized treatments. These treatments vary over a broad spectrum from immunization with patient-specific purified cancer antigens, to the induction of anti-tumor immunity with irradiated, patient-derived, cancer cells. Cell-based therapies can be further specialized by transfection with additional immune stimulatory molecules.Citation18 These autologous immunotherapies contain a wide array of proteins and peptides specific to the individual’s tumor antigens presented by self major histocompatibility complex (MHC) to stimulate the immune system directly.Citation19 Moreover, recent studies have demonstrated that tumors are made up of a highly heterogeneous population of cancer cells.Citation20,Citation21 Therefore, it can be hypothesized that autologous cell lines may be insufficient in the elicitation of a broad immune response and an entire primary tumor mass may be required for effective vaccination.

Allogeneic vaccines are largely similar to autologous vaccines with the exception that material is sourced from another member of the same species. Commonly used allogeneic materials include use of established laboratory-grown cancer cell lines known to express TAAs of a specific tumor type. This allows these therapeutics to be mass-produced, stored, and modified prior to use.Citation22 Although many allogeneic cell based vaccines are from cancer cell lines of the same species and type (breast, prostate, lung, etc.), they may differ from autologous vaccines in that they do not contain “patient-specific” tumor antigens.Citation23 Despite this disadvantage, preclinical studies in prostate cancerCitation24 and melanomaCitation25 models have demonstrated an added benefit of allogeneic sourced vaccines; presentation of TAAs in an allogeneic setting increases immunogenicity. Although, the supporting data are largely preclinical,Citation24 the hypothesis that allogeneic cells provide an additional danger signal is consistent with increased graft vs. tumor responses observed in patients who receive mismatched minor histocompatibility antigen bone marrow transplants.Citation26 Moreover, other desirable properties including availability, low production cost, and the lack of invasive procedures make allogeneic immunotherapy a salient modality.Citation27

Allogeneic Tumor-Based Vaccination Strategies

Allogeneic or autologous tumor vaccines can be prepared and administered to patients using irradiated whole cells or cell lysates. The use of these vaccine modalities allow for a reproducible, safe vaccine product in which injected tumor cells cannot replicate.Citation28 Additionally, irradiated cells naturally express and present numerous TAAs, alleviating the need to identify and purify TAAs, and thus promoting the initiation of an anti-tumor specific immune response.Citation19,Citation29 It has also been shown that immune recognition of tumor cells by CD8+T cells and antigen-presenting cells (APCs) is enhanced following irradiation.Citation30,Citation31 Furthermore, whole cell vaccines can be modified to increase immunogenicity by transfection of immune stimulatory molecules ex vivo.Citation32,Citation33 For example, the GVAX vaccine platform involves the use of irradiated allogeneic tumor cell lines, modified to secrete the cytokine granulocyte-macrophage colony-stimulating factor (GM-CSF) to augment the immunogenicity of the vaccine.Citation32,Citation34

Despite these advantages, drawbacks of using irradiated whole tumor cells as a vaccine also exist. For example, upon irradiation of cells, phosphatidylserine (PS), an immunosuppressive phospholipid usually found on the inner leaflet of the plasma membrane, is transferred to the outer leaflet. Expression of PS on the cell surface of irradiated cells has been shown to induce secretion of immunosuppressive factors by dendritic cells (DC) and inhibit maturation, promoting an immunosuppressive tumor environment. Furthermore, irradiated cells may retain the ability to secrete immunosuppressive factors like the original tumor cells.Citation35,Citation36 Thus irradiation induced immune suppression, partially negates the immunogenic effect of irradiated whole cells.Citation37 To overcome the immunosuppressive nature of irradiated whole tumor cell vaccines, Huang et al. generated anti-PS antibody:IL-2 immunocytokine complexes. The role of PS in immunosuppression and the ability of this biomolecule to block PS were confirmed by increased vaccine-specific immunogenicity in mice vaccinated with irradiated cells.Citation37,Citation38

Tumor cell lysates have also been used as vaccines to stimulate anti-tumor immune responses.Citation39 Although these vaccines allow for the presentation of multiple TAAs, they exhibit poor immunogenicity and some have been shown to be completely ineffective. The inability of tumor cell lysates to stimulate a sustained immune response may be due in part to the presence of immune suppressive molecules within the lysate.Citation40 To offset these deficiencies, many attempts have been made to enhance immunogenicity through the administration of adjuvant along with the vaccines. For example, Dong et al., showed that tumor cell lysates from Lewis lung cancer cells mixed with mycobacterial heat shock protein (HSP)65 were able to inhibit tumor growth and prolong the survival of lung cancer bearing mice by the activation of tumor specific cytotoxic T lymphocytes (CTLs) as well as innate immune cells.Citation41

Another whole cell lysate vaccination strategy utilizes DCs and their ability to efficiently capture and present antigen to T cells. This method, known as DC pulsing, allows for the activation of tumor specific CD4+ and CD8+ T cells through the MHC:peptide TCR interaction.Citation42,Citation43 DCs serve as an ideal link to the adaptive immune system due to high expression of MHC class I and II and costimulatory molecules and the subsequent initiation of an immune response. Moreover, DCs are also able to cross present exogenously loaded antigen and induce CD8+ T cell responses. These characteristics make DCs ideal for ex vivo loading of tumor cell lysates as evidenced by clinical studies in which administration of DC pulsed vaccines generates a large repertoire of tumor-specific immune responsesCitation44,Citation45

Clinical Trials with Allogeneic Cancer Vaccines

The following section provides a comprehensive look at results from clinical trials for various cancers in which disease stabilization is often achieved in conjunction with vaccine-induced immune responses. The results of these studies are summarized in .

Table 1. Overview of findings from allogeneic clinical trials for cancer immunotherapies

Breast Cancer

Due to the lack of effective treatment options for patients with late stage or metastatic breast cancerCitation46 and the difficulty of culturing primary cancer cells, allogeneic whole cell vaccines are currently being investigated as a potential adjuvant therapy to chemotherapy. Vaccines consisting of genetically modified tumor cells secreting various cytokines, such as GM-CSF, have previously been shown to be safe and induce an immune response in phase I and phase II clinical trials.Citation47 However, these responses were not sufficient to overcome the immune suppression induced by established tumors. In order to be effective as a monotherapy, cancer vaccines must be capable of inducing potent and sustainable tumor-specific immune responses that will reduce overall tumor burden. This provides a growing precedent for effectively combining immunotherapies with cytotoxic agents.Citation14

In a phase I clinical trial enrolling 28 patients with metastatic breast cancer, the safety and clinical efficacy of an allogeneic GM-CSF-secreting breast cancer vaccine was investigated. The vaccine, formulated from two HER2/neu positive mammary adenocarcinoma breast cancer cell lines (SKBR3 and T47D)Citation48 were administered either alone or in sequence with common chemotherapeutic agents cyclophosphamide (CY) and doxorubicin (DOX).Citation47 The goals of the study were to determine the safety of this combinatorial therapy and the optimal chemotherapy dose that would induce enhanced immunity against the breast cancer TAA HER2.Citation49

This study showed that up to 4 doses were well tolerated by patients, while accompanied by only modest levels of toxicity that was not exacerbated with the addition of chemotherapy. Additionally, the vaccine either alone or in sequence with low-dose chemotherapy (200 mg/m2 CY) could induce HER2-specific T-cell mediated immunity. HER2-specific humoral immunity was enhanced in the serum of patients receiving the vaccine along with 200 mg/m2 CY or 35 mg/m2 DOX.Citation47

These results suggest that low dose chemotherapy can be used to break tolerance, while sustaining an antigen-specific immune response. Moreover, these results are consistent with a working hypothesis that low dose chemotherapy can delay the vaccine specific immune response witnessed after repeated vaccination with allogeneic cells. When taken with preclinical data that DOXCitation14 and CYCitation50 augment anti-tumor immune responses while depleting Tregs,Citation51 a combinatorial approach of chemotherapy and immunotherapy should be considered strongly as a multivalent therapeutic modality.

In an another phase I study, the costimulatory molecule B7-1 (CD80) was transfected into HLA-A2+ matched allogeneic MDA-MB-231 breast cancer cell line and used as a vaccine to treat stage IV breast cancer patients.Citation52 Although vaccinated patients showed an increased tumor-specific immune response there was no tumor regression. However, the administration of a combination of allogeneic breast cancer cells (MCF-7), autologous breast cancer cells, TAA and low doses of systemic GM-CSF and IL-2 showed both a significant increase in antigen-specific lymphocyte response and an improved clinical response.Citation53 A separate study also reported that a similar multi-antigen combination vaccination consisting of TAAs, allogeneic, and autologous cells with systemic administration of biological adjuvants improved 10-y survival of breast cancer patients significantly in patients with depressed lymphocyte immunity at the start of the treatment (59% for unvaccinated group compared with 89% vaccinated group).Citation54 These observations suggest that a multi-antigen vaccination approach in combination with biological adjuvants is another promising therapeutic approach for breast cancer.

Lung Cancer

Counteracting the immunosuppressive environment created by primary tumors, such as those formed in non-small cell lung cancers (NSCLC), presents an opportunity for immune mediated disease stabilization. Recent developments including the identification of lung cancer TAAs (), immunosuppressive factors, and the establishment of multiple tumor cell lines have contributed to the growing number of available reagents.Citation55 The immunotherapeutic Belagenpumatucel-L (Lucanix), which is currently undergoing randomized phase III, utilizes multiple allogeneic tumor cell lines. Results from the prior phase II clinical trial conducted by Nemunaitis et al. demonstrated that subgroup analysis of individuals with anti-vaccine immune responses had an increased overall survival (OS).Citation56 Enrolling 75 patients with both early and late stage (stage II, IIIA, IIIB, and IV) NSCLC, patients were vaccinated with Lucanix, a cocktail of two adenocarcinomas, one squamous carcinoma, and one large-cell carcinoma cell lines (NCI-H-460, NCI-H-520, SK-LU-1, and Rh-2) transfected with a TGF- β2 antisense transgene to knockdown TGF-β2. Because TGF- β2 is a cytokine known to mediate immunosuppression high levels and correlate with poor prognosis, knock down of TGF- β2 could activate an anti-tumor immune response. This trial resulted in a one-year survival rate of 68% and a two-year survival rate of 52% for patients receiving the higher vaccine dose and 39% and 20%, respectively, for patients receiving a lower dose. Serum cytokine analysis determined significantly increased levels of IFN-γ, IL-6, and IL-4 in patients achieving disease stabilization compared with patients with progressive disease. Furthermore, patients were able to generate a positive ELISPOT response when stimulated with Lucanix specific antigen after vaccination.Citation57 These results indicated that favorable clinical response correlates with vaccine-induced immune responses justified further clinical investigation.

Table 2. Known TAAs of various cancers

The expression of costimulatory molecules is thought to increase the potency of allogeneic vaccines and direct the immune response. A phase I study enrolling 19 patients with advanced stage (stage IIIB, IV) metastatic NSCLC employed this strategy. Patients were vaccinated with an irradiated whole cell allogeneic vaccine (cell line AD100) transfected to express the costimulatory molecule B7-1 along with either HLA-A1 or HLA-A2.Citation55 The costimulatory molecule B7-1 was administered to boost the expansion of tumor specific immune responses that may help overcome the immunosuppressive environment present in NLSC. The patients were administered a vaccine depending on their HLA type; A1 patients received the A1 vaccine for direct antigen presentation, and non-A1 and non-A2 patients, received an unmatched vaccine (A1) for indirect antigen presentation. The median survival was found to be 18 mo with 32% of the patients achieving disease stabilization. After subgroup analysis, the authors found that disease stabilization was not HLA restricted, suggesting that indirect presentation not only sustained an anti-tumor response, but also enhanced it through allogeneic MHC molecules. Furthermore, an ELISPOT assay using CD8+ T-cells isolated from patients after treatment showed increased IFN-γ production by vaccine stimulated CD8+ T cells when compared with levels before treatment.Citation58 The ability of this allogeneic vaccine to stimulate a tumor specific CD8+ T-cell response and increase patient survival in patients with advanced disease bodes well for cancer immunotherapies.Citation55,Citation58

Recently, results from a novel immunotherapeutic approach for the treatment of NSCLC indicate that allogeneic tumor cell lines secreting HSPgp96-Ig show that peptides from allogeneic tumor cells delivered to patients’ immune system via gp96 can induce an immune response.Citation59 While no objective tumor response was observed in clinical trials, stabilization of tumor growth and increased release of IFN-γ by CD8+ T Cells warrant further exploration of similar approaches.

Leukemia

Although chemotherapy, radiation therapy and transplantation have become the standard of care in leukemia, a study was conducted to test the efficacy of an allogeneic tumor lysate pulsed DC based vaccine in patients with B-cell chronic lymphoma leukemia (B-CLL).Citation60 The therapeutic regimen involved five intradermal administrations of DCs pulsed ex vivo with allogeneic tumor lysate or apoptotic bodies. Of the 9 patients enrolled in the study, 4 patients achieved disease stabilization for over 23 mo as determined by stabilized white blood cell and lymphocyte counts. However, only 1 of the 4 HLA matched patients was able to raise a detectable anti-TAA immune response. While this phase I trial shows that the therapy was tolerable and had positive immune correlates, immunotherapies in leukemia may provide maximum therapeutic benefit as an adjuvant therapy after reduction of disease burden through ablative therapies and transplantation. Thus, optimizing dosage of myeloablative drugs and immunotherapeutics may provide the greatest clinical benefit.

Chronic myeloid leukemia (CML) is another type of leukemia that responds well to chemotherapeutic drugs. However, the residual disease persists following treatment. A clinical trial was conducted to determine whether vaccination with GM-CSF-secreting K562 cells could induce an antitumor immune response in CML patients receiving the chemotherapeutic drug imatinib mesylate.Citation61 The K562 cell line was chosen for this immunotherapy, as it is an established human CML cell line that expresses many CML antigens. Results from this clinical trial with 19 patients determined that vaccination with GM-CSF secreting K562 cells was capable of diminishing disease below detection levels in seven patients and lowered tumor burden in 12 patients. These results further emphasize the role of the immune system in eliminating and controlling the growth of residual cancer cells resistant to standard chemotherapies and the efficacy of immunotherapies when used in an adjuvant fashion.

Melanoma

The most notable melanoma immunotherapeutic to date, Canavaxin-polyvalent (PV), was tested at the John Wayne Cancer Institute in the largest single institution stage-IIICitation62 melanoma trial ever conducted. In the phase II trial, the allogeneic polyvalent whole cell vaccine formulation Canavaxin-PV, was demonstrated to be an effective adjuvant and immunotherapy. The vaccine regimen consisted of intradermal injections for 1 y following lymphadenectomy, and resulted in a significant increase in 5 y OS compared matched, untreated groups. However, it seems that the subgroup analysis, which identified Canavaxin-PV’s efficacy, did not correctly identify factors correlating to vaccine efficacy. The two subsequent randomized phase III trials testing Canvaxin-PV + the adjuvant BCG vs. placebo + BCG in stage-III and stage-IV melanoma were halted before completion, because no beneficial effect between the vaccine treated and the placebo treated group was observed.

While the reason for failure of this trial is uncertain, Canavaxin-PV had many characteristics needed for an allogeneic cell based vaccine. This formulation consisted of 3 melanoma cell lines (M10-V, M24-V, and M101-V) and contained over 20 distinct melanoma-associated antigens. Moreover, immunologic analyses of the phase II trial demonstrated a correlation between matched MHC haplotype and increased OS. This vaccine regimen, however, could be augmented by cytokine stimulation and the use of another adjuvant. This trial underscores the necessity for a multifactorial approach for effective immune activation in cancer patients.

Other treatment strategies for melanoma have focused on presentation of a number of TAAs through other methods. Established through a series of clinical trials, Melacine® has exhibited the effectiveness of lysate based vaccines in the melanoma model.Citation63 The allogeneic melanoma cell lysate, utilized cell lines (Mel-D and Mel-S) isolated from biopsies of patients with nodular melanoma.Citation64 Administered as the lysate of multiple allogeneic cell lines with the adjuvant detoxified endotoxin and mycobacterium cytoskeleton (DETOX), this vaccine increased melanoma specific CTL precursors, increased cancer cell specific antibody titers, and induced immunity as evidenced by delayed-type hypersensitivity reactions in response to autologous cancer cells. In phase I clinical trials this vaccination regimen was able to induce remission in 5 out of 17 patients (29%).

Subsequent phase II and phase III clinical trials did not show such promising results, however, with only 5 complete remissions and 7 partial remissions out of 129 patients (overall objective response rate 6.1%). In an attempt to supplement the vaccination strategy, Melacine® was administered along with a single dose of 300 mg/m2 CY and IFN-α intravenously after 4 doses of Melacine®. Of the 39 patients that were evaluated, there was only an overall objective response of 10%. Nevertheless, 64% of the patients achieved disease stabilization for 16 weeks.Citation65 Further investigation confirmed the induction of an anti-vaccine immune response against multiple melanoma antigens including Tyrosinase, MART-1, gp100, MAGE-1, MAGE-2, and MAGE-3.Citation66 The results of this trial augment a growing body of literature supporting the use of immunotherapies in a minimal residual disease setting.

While similar strategies such as immunization with recombinant MART-1 peptides have also been shown some efficacy, whole cell vaccines and cell lysates have the added advantage of stimulating T cells with unidentified intracellular and extracellular melanoma antigens.Citation67 Loading these antigens onto DCs confers the additional ability to directly prime CD8+ CTLs through cross presentation of exogenously acquired antigen. The efficacy of DCs to induce anti-tumor immunity was demonstrated in phase I clinical trials. Patients received autologous DCs loaded ex vivo with antigens from apoptotic/necrotic allogeneic melanoma cells. Anti-tumor immunity was confirmed by the induction of gp100 and MelanA/MART-1-specific CTLs.Citation68

This study supports data presented by Palucka et al. in 2006.Citation69 In this study, patients with stage-IV melanoma were vaccinated with autologous monocyte derived DCs loaded with killed allogeneic Colo829 melanoma cells and activated with GM-CSF, IL-4, TNF-α and CD40 ligand. The study participants had a median OS of 22.5 mo in conjunction with increased IFN-γ secretion by lymphocytes and proliferation in the presence of a Colo829 TAA peptide. CD8+ T-cell immunity to novel peptides was also observed, indicating that DC cross presentation had occurred.

The relative success of this phase I trial compared with allogeneic DC based vaccinations can be attributed to its multifactorial approach. The use of primed autologous DCs loaded with allogeneic TAAs from the Colo829 cell line, addressed problems concerning HLA matched presentation of antigen and DC anergy. This vaccination strategy demonstrates the importance of utilizing both the innate immune system and TAAs in eliciting an effective anti-tumor immune response.

Taken together, observations from these clinical trials in melanoma seem to indicate that polyvalent strategies that augment a sufficient immune response need to be considered.

Prostate Cancer

The development of an effective immunotherapy to treat prostate cancer has seen modest success in recent years. The immunotherapy Onyvax-P consists of three irradiated prostate cancer cell lines (OnyCap23, LnCaP, and P4E6) representative of three different stages of prostate cancer. The vaccine formulation was administered with the adjuvant BCG to individuals with asymptomatic hormone-resistant prostate cancer (HRPC). Among the 26 patients with increased levels of PSA enrolled in the study, administration of the vaccine resulted in decreased PSA velocity (PSAV) in 42% of the patients and increased median time to disease progression from 28 weeks to 58 weeks.Citation70 Immunological studies indicated that patients with decreased PSAV showed a Th1 cytokine release pattern when T cells were stimulated by lysate ex-vivo compared with patients who did not respond to the vaccine and exhibited both a Th1 and Th2 cytokine release patterns.

The subsequent phase I/II clinical trial explored the administration of a whole cell vaccine consisting of a mixture of three proprietary (Onyvax Ltd.) prostate cancer cell linesCitation71 along with the bacterial adjuvant BCG.Citation72 Throughout the course of the study, levels of PSA, cytokines, prostate-specific T cells, and serum antibody titer were measured. Although small decreases in PSA could not be attributed solely to the vaccine, increased levels of cytokine production and prostate-specific antibody titer indicated that the vaccination led to a specific immune response. This series of clinical trials highlights the importance of disease burden on the efficacy of an allogeneic tumor immunotherapy approach. The initial vaccine trial for Onyvax-P enrolled patients with advanced HRPC and showed limited efficacy. However, the successful treatment of patients with asymptomatic HRPC in this study indicates the limitation of solely administering immunotherapies to patients with high disease burdens.

A cytokine secreting allogeneic modality was tested in prostate cancer by the GVAX platform. The regimen consisted of two allogeneic cell lines (PC3, LNCaP) modified to secrete GM-CSF.Citation73 PC3 is derived from prostate cancer bone marrow metastases and is castration resistant.Citation74 The LNCaP cell line derived from prostate cancer lymph node metastases is hormone sensitive, and expresses numerous prostate-associated antigens.Citation75 Together, these two cells lines provide a broad range of antigens that are prevalent in various prostate cancers. Preclinical data showed that GVAX was successful in inducing anti-tumor immunity in melanoma, lymphoma, colon, fibrosarcoma, lung, renal, and prostate cancers. A potential mechanism for this anti-tumor response is the concurrent activation of APCs and arms of the adaptive immune system.Citation76 Phase I/II clinical trials testing the efficacy and safety of GVAX as a monotherapy, enrolled 34 patients with metastatic castration resistant prostate cancer (CRPC). The trial demonstrated that patients receiving the vaccine had a median OS of 26.2 mo, comparable to patients that receive chemotherapy.Citation77 Based on the findings of these small phase I/II trials, two large Phase III randomized studies (VITAL-1 and VITAL-2) were developed to test the clinical efficacy of GVAX in treating metastatic prostate cancer.Citation78

Production of GVAX was halted after both VITAL-1 and VITAL-2 were terminated due to the inefficacy of GVAX and excessive deaths, respectively.Citation79 Understanding the failure of GVAX in metastatic prostate cancer would provide useful information toward the improvement of this vaccination and similar modalities.

To test GVAX in a combinatorial setting, a clinical trial was performed in which both GVAX and Ipilimumab were administered in escalating doses.Citation80 Ipilimumab is a fully human IgG CTLA-4 blocking antibody. CTLA-4 is a homolog of CD28 found on T cells generally 2–3 d post activation. Its ability to bind B7-1 and B7-2 with a 100-fold greater affinity than CD28 allows it to compete with CD28 for B7-1 and B7-2 binding, thus terminating the T cell response and limiting the T cell population pool size.Citation80 The study showed disease regression as determined by reduction of PSA levels in 5 of 12 patients diagnosed with metastatic HRPC. This correlated directly with Ipilimumab dosage as 5 of the 6 patients received a higher dosage of Ipilimumab.Citation81 Although GVAX was more efficacious as a combinatorial treatment, patients faced several endocrine complications at higher dosages indicating the need for more specific targeting mechanisms if such combinatorial therapies are to be employed.

Similar to GVAX, the LNCaP cell line was modified using a retroviral vector to secrete IL-2 and IFN-γ.Citation82 Six patients with HRPC were enrolled in the study. During the course of the study serum PSA levels and CTL generation were monitored. A 2-fold higher response to various antigens was seen in all patients. Interestingly, an inverse correlation was observed between levels of serum PSA and antigen specific immune cells in circulation. These results support the hypothesis that T cells migrate from the periphery into vascular circulation to mediate an immune response.

As these clinical trials in prostate cancer have demonstrated, a vaccine response can vary widely based on cytokine costimulation, allogeneic cell lines administered and vaccine modality. Moreover, these trials also identify disease burden as a significant factor in the efficacy of an immunotherapy during clinical evaluation. It is thus critical to determine the appropriate clinical settings in which these immunotherapies are most effective as a monotherapy, combinatorial therapy, or as an adjuvant therapy.

Pancreatic Cancer

Since 1975, the rate of incidence of pancreatic cancer has been increasing by 1.5% per year, a figure attributed partly to the lack of a widespread detection method.Citation83 Although TAAs () have been identified for pancreatic cancer, correlation between detection of these antigens and disease prognosis remain highly unreliable. This lack of knowledge justifies the exploration of allogeneic whole cell vaccines as immunotherapies for patients with pancreatic cancer.Citation84

Currently, the standard of care for advanced pancreatic cancer is the chemotherapeutic drug gemcitabine.Citation85 Clinical trials of allogeneic vaccines for pancreatic cancer are currently underway and the early phase of trials have reported promising results. Two allogeneic vaccines in currently tested in clinical trials for the treatment of pancreatic cancer include NewLink Genetic’s HyperAcute® pancreatic cancer vaccine (Algenpantucel-L) and BioSante Pharmaceutical’s GVAX Pancreas Vaccine. Algenpantucel-L (HyperAcute-Pancreas) combines two human allogeneic pancreatic cancer cell lines expressing α-1,3-galactosyltransferase, a murine enzyme that mediates α-galactosyl (αGal) epitope expression on the surface of the cells. The rationale behind this vaccine strategy is to take advantage of naturally produced, anti-αGal antibodies by the human body as an adjuvant.Citation86 The anti-tumor immune response resulting through opsonization, complement activation, and ADCC of anti-αGal antibody:αGal immune complexes.Citation87,Citation88 The phase II clinical trial for HyperAcute-Pancreas generated favorable results with 62% of patients remaining disease-free at the one-year endpoint, and an overall survival of 24.1 mo.Citation89 A phase III clinical trial, which began in 2010, enrolled 722 patients with stage-I and stage-II pancreatic cancer to study the effect HyperAcute-Pancreas in combination with chemotherapy or chemoradiotherapy.

The previously discussed GVAX vaccine platform has also been tested in pancreatic cancer. In an initial 14-patient phase I clinical trial testing for the safety of the GVAX regimen, Jaffe et al. reported that 3 of 14 patients were disease-free for at least 25 mo after treatment.Citation90 Combinatorial approaches employing GM-CSF secreting cell lines and chemotherapy are also being explored in metastatic pancreatic cancer. In 2008, a phase I clinical trial purported minimal toxicity and enhanced T cell function in patients receiving an immunotherapy and cyclophosphamide.Citation91 These findings suggest chemotherapeutic treatments can enhance the activity of an immunotherapy as exhibited by higher rates of antigen-specific CD8+ T-cell activation.

Another clinical trial for a GM-CSF secreting immunotherapy also recently explored the combinatorial use of immunotherapy, radiotherapy and chemotherapy as an adjuvant, post resection of pancreatic adenocarcinoma.Citation92 This single arm phase II trial demonstrated that administration of up to 5 doses of the vaccine induced antigen-specific T cells correlating with increased overall survival. Patients that remained disease free generated lymphocytes that could respond to a greater variety of antigen after receiving the combinatorial therapy. The results of these trials provide a strong foundation for growing evidence that immunotherapies should be considered for use as an adjuvant therapy to resection or in a combination with established chemotherapeutic regimen in the treatment of pancreatic cancer.

Other Cancers

The CancerVax regimen designed initially for the treatment of melanoma, consists of three live-irradiated melanoma cell lines that include many immunogenic colon carcinoma TAAs including the glycoprotein TA90. A phase I clinical trial was conducted to determine whether administration of CancerVax and the adjuvant BCG could induce a vaccine-specific immune response which correlated with overall survival in patients with advanced colon cancer.Citation93 The trials results indicated that vaccination increased median OS compared with historical controls and that patients with high titers of TA90-IgM immune complexes had a significantly higher OS. This trial demonstrates the potential for allogeneic therapies to treat a broad array of cancers given a set of common TAAs.

In a clinical trial for metastatic renal cancer, 10 HLA-A-0201 matched patients were enrolled to test the efficacy of an HLA matched allogeneic renal cancer cell line (RCC26) transfected with IL-7 and B7-1.Citation94 Despite an increase in median OS, immune correlates suggested that vaccination induced a Th2-polarized response marked by release of IL-10 upon stimulation with vaccine-specific peptide. Furthermore, the vaccine failed to rescue tolerized lymphocytes, which were incapable of interferon production both in the presence of antigen-specific and non-specific stimulation. This implies that when employed as a monotherapy, the vaccine was not able to break tolerance. Therefore, this therapy must be employed either, concomitantly to deplete Tregs and break tumor immune suppression, or in a minimal residual disease setting.

The Future of Therapeutic Allogeneic Cancer Vaccines

These clinical trials demonstrate the ability of cell based allogeneic immunotherapies to stimulate a clinically relevant response. The presence of a tumor mass within an established immunosuppressive microenvironment is often an insurmountable barrier, for the immune system to overcome. As demonstrated by the vaccination of individuals with resected lung cancer and asymptomatic HRPC, the use of immunotherapies in a clinical setting either post surgery or post chemotherapy allows the immune system to respond more efficiently by killing residual tumors that escape initial treatment.Citation14 Furthermore phase I studies, which are performed on patients with advanced stage cancers with a poor prognosis provide a suboptimal environment for the assessment of these immunotherapies. Complicating these results further, patients participating in many clinical trials are often severely immunocompromised due to prior participation in various chemo and radiation therapies. With these limitations of the clinical trials process, allogeneic cancer vaccines could show the most clinical benefit when used as an adjuvant therapy.Citation95

It is also imperative to note the correlation between the effectiveness of an allogeneic vaccine and the number of common TAAs () expressed by both the cancer and the allogeneic cell line. The discovery of multiple TAAs that serve as excellent biomarkers for the disease allows not only for easy monitoring, but also for better engineering of cancer cell lines and the induction of anti-tumor specific immunity. This has been well documented in the literature, as vaccination with a greater number of allogeneic cell lines has proven more effective.

A major impediment to the use of allogeneic therapies, however, is the extensive intratumoral heterogeneity in cancers.Citation96 Through multi region tumor sequencing, one study has determined that between 63% and 69% of all somatic mutations were heterogeneous.Citation20 Additionally, the study enabled the construction of gene phylogenies, which unveiled the progressive loss of tumor-suppressor genes within a single tumor, indicating the convergence of tumor phenotype through multiple spatially separated mutations.Citation20 This line of evidence supports a model in which following the initial mutations needed for tumor escape, subclones are free to mutate divergently in a deregulated manner.Citation97 Moreover, the addition of selective pressures through the use of allogeneic immunotherapies may promote escape and drive further mutation and divergence. Thus it can be hypothesized that allogeneic tumor cell vaccines developed from established cell lines may not represent the antigenic characteristics of the entire tumor and therefore will face increasing levels of intratumoral complexity depending on the tumor progression within a given patient. Despite the induction of measurable antitumor immunity in many patients, the lack of successful remission or significant clinical benefit of allogeneic tumor immunotherapies when employed as a monotherapy may be explained by the pre-existing inter- and intra-tumoral heterogeneity and/or tumor induced immunosuppression at the microenvironment. In addition to the challenge posed by the inter- and intra-tumoral heterogeneity, many of the allogeneic tumor cell vaccine clinical trials discussed above did not incorporate strategies that will mitigate the tumor-induced immunosuppression. Therefore, in the future, a better therapeutic efficacy is expected when allogeneic tumor vaccines are developed based on the genomic and transcriptomic analysis of patient’s cancersCitation98 and administered in conjunction with anti-immunosuppressive agents.

Abbreviations:
APC=

antigen presenting cell

B-CLL=

B-cell chronic lymphoma leukemia

CIK=

cytokine induced killer

CRPC=

castration resistant prostate cancer

HSP=

heat shock protein

CTLs=

cytotoxic T lymphocytes

CY=

cyclophosphamide

DC=

dendritic cell

DOX=

doxorubicin

GM-CSF=

granulocyte macrophage-colony stimulating factor

GVHD=

graft versus host disease

GVL=

graft versus leukemia

HRPC=

hormone-refractory prostate cancer

MHC=

major histocompatibility complex

NK=

natural killer

NSCLC=

non-small cell lung cancers

OS=

overall survival

PFS=

progression free survival

PS=

phosphatidyl serine

PSA=

prostate-specific antigen

TAAs=

tumor-associated antigens

Disclosure of Potential Conflicts of Interest

No potential conflicts of interest were disclosed.

Acknowledgments

This work was supported by NIH grants R01 CA138993 (to Selvaraj P), F31 CA165632 (to Patel JM), and F31 CA165897 (to Bozeman EN). The authors thank Archana V Boopathy for critical reading of the manuscript.

10.4161/hv.26568

References

  • Sherr CJ. Cancer cell cycles. Science 1996; 274:1672 - 7; http://dx.doi.org/10.1126/science.274.5293.1672; PMID: 8939849
  • Malumbres M, Barbacid M. To cycle or not to cycle: a critical decision in cancer. Nat Rev Cancer 2001; 1:222 - 31; http://dx.doi.org/10.1038/35106065; PMID: 11902577
  • Evan GI, Vousden KH. Proliferation, cell cycle and apoptosis in cancer. Nature 2001; 411:342 - 8; http://dx.doi.org/10.1038/35077213; PMID: 11357141
  • Lowe SW, Cepero E, Evan G. Intrinsic tumour suppression. Nature 2004; 432:307 - 15; http://dx.doi.org/10.1038/nature03098; PMID: 15549092
  • Dunn GP, Old LJ, Schreiber RD. The immunobiology of cancer immunosurveillance and immunoediting. Immunity 2004; 21:137 - 48; http://dx.doi.org/10.1016/j.immuni.2004.07.017; PMID: 15308095
  • Kim R, Emi M, Tanabe K. Cancer immunoediting from immune surveillance to immune escape. Immunology 2007; 121:1 - 14; http://dx.doi.org/10.1111/j.1365-2567.2007.02587.x; PMID: 17386080
  • Swann JB, Smyth MJ. Immune surveillance of tumors. J Clin Invest 2007; 117:1137 - 46; http://dx.doi.org/10.1172/JCI31405; PMID: 17476343
  • Garrido F, Algarra I. MHC antigens and tumor escape from immune surveillance. Adv Cancer Res 2001; 83:117 - 58; http://dx.doi.org/10.1016/S0065-230X(01)83005-0; PMID: 11665717
  • Smyth MJ, Hayakawa Y, Takeda K, Yagita H. New aspects of natural-killer-cell surveillance and therapy of cancer. Nat Rev Cancer 2002; 2:850 - 61; http://dx.doi.org/10.1038/nrc928; PMID: 12415255
  • Ostrand-Rosenberg S. Immune surveillance: a balance between protumor and antitumor immunity. Curr Opin Genet Dev 2008; 18:11 - 8; http://dx.doi.org/10.1016/j.gde.2007.12.007; PMID: 18308558
  • Vallejo R, Hord ED, Barna SA, Santiago-Palma J, Ahmed S. Perioperative immunosuppression in cancer patients. J Environ Pathol Toxicol Oncol 2003; 22:139 - 46; http://dx.doi.org/10.1615/JEnvPathToxOncol.v22.i2.70; PMID: 14533877
  • Cavallo F, De Giovanni C, Nanni P, Forni G, Lollini PL. 2011: the immune hallmarks of cancer. Cancer Immunol Immunother 2011; 60:319 - 26; http://dx.doi.org/10.1007/s00262-010-0968-0; PMID: 21267721
  • Ringdén O, Le Blanc K. Allogeneic hematopoietic stem cell transplantation: state of the art and new perspectives. APMIS 2005; 113:813 - 30; http://dx.doi.org/10.1111/j.1600-0463.2005.apm_336.x; PMID: 16480452
  • Emens LA, Jaffee EM. Leveraging the activity of tumor vaccines with cytotoxic chemotherapy. Cancer Res 2005; 65:8059 - 64; http://dx.doi.org/10.1158/0008-5472.CAN-05-1797; PMID: 16166275
  • Mantovani A, Schioppa T, Porta C, Allavena P, Sica A. Role of tumor-associated macrophages in tumor progression and invasion. Cancer Metastasis Rev 2006; 25:315 - 22; http://dx.doi.org/10.1007/s10555-006-9001-7; PMID: 16967326
  • Melcher A, Todryk S, Hardwick N, Ford M, Jacobson M, Vile RG. Tumor immunogenicity is determined by the mechanism of cell death via induction of heat shock protein expression. Nat Med 1998; 4:581 - 7; http://dx.doi.org/10.1038/nm0598-581; PMID: 9585232
  • Tan T-T, Coussens LM. Humoral immunity, inflammation and cancer. Curr Opin Immunol 2007; 19:209 - 16; http://dx.doi.org/10.1016/j.coi.2007.01.001; PMID: 17276050
  • Colombo MP, Forni G. Cytokine gene transfer in tumor inhibition and tumor therapy: where are we now?. Immunol Today 1994; 15:48 - 51; http://dx.doi.org/10.1016/0167-5699(94)90131-7; PMID: 8155261
  • Moingeon P. Cancer vaccines. Vaccine 2001; 19:1305 - 26; http://dx.doi.org/10.1016/S0264-410X(00)00372-8; PMID: 11163653
  • Gerlinger M, Rowan AJ, Horswell S, Larkin J, Endesfelder D, Gronroos E, Martinez P, Matthews N, Stewart A, Tarpey P, et al. Intratumor heterogeneity and branched evolution revealed by multiregion sequencing. N Engl J Med 2012; 366:883 - 92; http://dx.doi.org/10.1056/NEJMoa1113205; PMID: 22397650
  • Russnes HG, Navin N, Hicks J, Borresen-Dale A-L. Insight into the heterogeneity of breast cancer through next-generation sequencing. J Clin Invest 2011; 121:3810 - 8; http://dx.doi.org/10.1172/JCI57088; PMID: 21965338
  • Copier J, Dalgleish A. Overview of tumor cell-based vaccines. Int Rev Immunol 2006; 25:297 - 319; http://dx.doi.org/10.1080/08830180600992472; PMID: 17169778
  • Ueda R, Shiku H, Pfreundschuh M, Takahashi T, Li LTC, Whitmore WF, Oettgen HF, Old LJ. Cell surface antigens of human renal cancer defined by autologous typing. J Exp Med 1979; 150:564 - 79; http://dx.doi.org/10.1084/jem.150.3.564; PMID: 479762
  • Hrouda D, Todryk SM, Perry MJA, Souberbielle BE, Kayaga J, Kirby RS, Dalgleish AG. Allogeneic whole-tumour cell vaccination in the rat model of prostate cancer. BJU Int 2000; 86:742 - 8; http://dx.doi.org/10.1046/j.1464-410x.2000.00887.x; PMID: 11069388
  • Knight BC, Souberbielle BE, Rizzardi GP, Ball SE, Dalgleish AG. Allogeneic murine melanoma cell vaccine: a model for the development of human allogeneic cancer vaccine. Melanoma Res 1996; 6:299 - 306; http://dx.doi.org/10.1097/00008390-199608000-00004; PMID: 8873049
  • Katagiri T, Shiobara S, Nakao S, Wakano M, Muranaka E, Kuba N, Furukawa T, Tsukada J, Takeda H, Aizawa Y, et al. Mismatch of minor histocompatibility antigen contributes to a graft-versus-leukemia effect rather than to acute GVHD, resulting in long-term survival after HLA-identical stem cell transplantation in Japan. Bone Marrow Transplant 2006; 38:681 - 6; http://dx.doi.org/10.1038/sj.bmt.1705506; PMID: 16980988
  • de Gruijl TD, van den Eertwegh AJM, Pinedo HM, Scheper RJ. Whole-cell cancer vaccination: from autologous to allogeneic tumor- and dendritic cell-based vaccines. Cancer Immunol Immunother 2008; 57:1569 - 77; http://dx.doi.org/10.1007/s00262-008-0536-z; PMID: 18523771
  • Deacon DH, Hogan KT, Swanson EM, Chianese-Bullock KA, Denlinger CE, Czarkowski AR, Schrecengost RS, Patterson JW, Teague MW, Slingluff CL Jr.. The use of gamma-irradiation and ultraviolet-irradiation in the preparation of human melanoma cells for use in autologous whole-cell vaccines. BMC Cancer 2008; 8:360; http://dx.doi.org/10.1186/1471-2407-8-360; PMID: 19055839
  • Li J, King AV, Stickel SL, Burgin KE, Zhang X, Wagner TE, Wei Y. Whole tumor cell vaccine with irradiated S180 cells as adjuvant. Vaccine 2009; 27:558 - 64; http://dx.doi.org/10.1016/j.vaccine.2008.11.014; PMID: 19027812
  • Sharma A, Bode B, Wenger RH, Lehmann K, Sartori AA, Moch H, Knuth A, Boehmer Lv, Broek Mv. γ-Radiation promotes immunological recognition of cancer cells through increased expression of cancer-testis antigens in vitro and in vivo. PLoS One 2011; 6:e28217; http://dx.doi.org/10.1371/journal.pone.0028217; PMID: 22140550
  • Obeid M, Panaretakis T, Joza N, Tufi R, Tesniere A, van Endert P, Zitvogel L, Kroemer G. Calreticulin exposure is required for the immunogenicity of gamma-irradiation and UVC light-induced apoptosis. Cell Death Differ 2007; 14:1848 - 50; http://dx.doi.org/10.1038/sj.cdd.4402201; PMID: 17657249
  • Simons JW, Sacks N. Granulocyte-macrophage colony-stimulating factor-transduced allogeneic cancer cellular immunotherapy: The GVAX (TM) vaccine for prostate cancer. Urologic Oncology-Seminars and Original Investigations 2006; 24:419 - 24; http://dx.doi.org/10.1016/j.urolonc.2005.08.021
  • Pardoll DM. Spinning molecular immunology into successful immunotherapy. Nat Rev Immunol 2002; 2:227 - 38; http://dx.doi.org/10.1038/nri774; PMID: 12001994
  • Parmiani G, Castelli C, Pilla L, Santinami M, Colombo MP, Rivoltini L. Opposite immune functions of GM-CSF administered as vaccine adjuvant in cancer patients. Ann Oncol 2007; 18:226 - 32; http://dx.doi.org/10.1093/annonc/mdl158; PMID: 17116643
  • Satoh E, Naganuma H, Sasaki A, Nagasaka M, Ogata H, Nukui H. Effect of irradiation on transforming growth factor-beta secretion by malignant glioma cells. J Neurooncol 1997; 33:195 - 200; http://dx.doi.org/10.1023/A:1005791621265; PMID: 9195490
  • Klopp AH, Spaeth EL, Dembinski JL, Woodward WA, Munshi A, Meyn RE, Cox JD, Andreeff M, Marini FC. Tumor irradiation increases the recruitment of circulating mesenchymal stem cells into the tumor microenvironment. Cancer Res 2007; 67:11687 - 95; http://dx.doi.org/10.1158/0008-5472.CAN-07-1406; PMID: 18089798
  • Huang X, Ye D, Thorpe PE. Enhancing the potency of a whole-cell breast cancer vaccine in mice with an antibody-IL-2 immunocytokine that targets exposed phosphatidylserine. Vaccine 2011; 29:4785 - 93; http://dx.doi.org/10.1016/j.vaccine.2011.04.082; PMID: 21557977
  • Huynh MLN, Fadok VA, Henson PM. Phosphatidylserine-dependent ingestion of apoptotic cells promotes TGF-beta1 secretion and the resolution of inflammation. J Clin Invest 2002; 109:41 - 50; PMID: 11781349
  • Zeng Y, Feng HP, Graner MW, Katsanis E. Tumor-derived, chaperone-rich cell lysate activates dendritic cells and elicits potent antitumor immunity. Blood 2003; 101:4485 - 91; http://dx.doi.org/10.1182/blood-2002-10-3108; PMID: 12576309
  • Graner MW, Likhacheva A, Davis J, Raymond A, Brandenberger J, Romanoski A, Thompson S, Akporiaye E, Katsanis E. Cargo from tumor-expressed albumin inhibits T-cell activation and responses. Cancer Res 2004; 64:8085 - 92; http://dx.doi.org/10.1158/0008-5472.CAN-04-1871; PMID: 15520220
  • Dong B, Sun L, Wu X, Zhang P, Wang L, Wei H, Zhou L, Hu X, Yu Y, Hua S, et al. Vaccination with TCL plus MHSP65 induces anti-lung cancer immunity in mice. Cancer Immunol Immunother 2010; 59:899 - 908; http://dx.doi.org/10.1007/s00262-010-0816-2; PMID: 20087582
  • Yu JS, Liu GT, Ying H, Yong WH, Black KL, Wheeler CJ. Vaccination with tumor lysate-pulsed dendritic cells elicits antigen-specific, cytotoxic T-cells in patients with malignant glioma. Cancer Res 2004; 64:4973 - 9; http://dx.doi.org/10.1158/0008-5472.CAN-03-3505; PMID: 15256471
  • Geiger JD, Hutchinson RJ, Hohenkirk LF, McKenna EA, Yanik GA, Levine JE, Chang AE, Braun TM, Mulé JJ. Vaccination of pediatric solid tumor patients with tumor lysate-pulsed dendritic cells can expand specific T cells and mediate tumor regression. Cancer Res 2001; 61:8513 - 9; PMID: 11731436
  • Höltl L, Zelle-Rieser C, Gander H, Papesh C, Ramoner R, Bartsch G, Rogatsch H, Barsoum AL, Coggin JH Jr., Thurnher M. Immunotherapy of metastatic renal cell carcinoma with tumor lysate-pulsed autologous dendritic cells. Clin Cancer Res 2002; 8:3369 - 76; PMID: 12429623
  • Pandha HS, John RJ, Hutchinson J, James N, Whelan M, Corbishley C, Dalgleish AG. Dendritic cell immunotherapy for urological cancers using cryopreserved allogeneic tumour lysate-pulsed cells: a phase I/II study. BJU Int 2004; 94:412 - 8; http://dx.doi.org/10.1111/j.1464-410X.2004.04922.x; PMID: 15291878
  • Curigliano G, Locatelli M, Fumagalli L, Goldhirsch A. Immunizing against breast cancer: a new swing for an old sword. Breast 2009; 18:Suppl 3 S51 - 4; http://dx.doi.org/10.1016/S0960-9776(09)70273-5; PMID: 19914543
  • Emens LA, Asquith JM, Leatherman JM, Kobrin BJ, Petrik S, Laiko M, Levi J, Daphtary MM, Biedrzycki B, Wolff AC, et al. Timed sequential treatment with cyclophosphamide, doxorubicin, and an allogeneic granulocyte-macrophage colony-stimulating factor-secreting breast tumor vaccine: a chemotherapy dose-ranging factorial study of safety and immune activation. J Clin Oncol 2009; 27:5911 - 8; http://dx.doi.org/10.1200/JCO.2009.23.3494; PMID: 19805669
  • Mehta RS, Schubbert T, Hsiang D, Butler J, Baick C, Su MY. . High pathological complete remission rates with paclitaxel and carboplatin +/− trastuzumab (TC +/− H) following dose dense doxorubicin and cyclophosphamide (AC) supported by GM-CSF in breast cancer-a phase I study. Breast Cancer Research and Treatment2005; 94:S225-S
  • Meden H, Kuhn W. Overexpression of the oncogene c-erbB-2 (HER2/neu) in ovarian cancer: a new prognostic factor. Eur J Obstet Gynecol Reprod Biol 1997; 71:173 - 9; http://dx.doi.org/10.1016/S0301-2115(96)02630-9; PMID: 9138962
  • Schiavoni G, Mattei F, Di Pucchio T, Santini SM, Bracci L, Belardelli F, Proietti E. Cyclophosphamide induces type I interferon and augments the number of CD44(hi) T lymphocytes in mice: implications for strategies of chemoimmunotherapy of cancer. Blood 2000; 95:2024 - 30; PMID: 10706870
  • Ercolini AM, Ladle BH, Manning EA, Pfannenstiel LW, Armstrong TD, Machiels JPH, Bieler JG, Emens LA, Reilly RT, Jaffee EM. Recruitment of latent pools of high-avidity CD8(+) T cells to the antitumor immune response. J Exp Med 2005; 201:1591 - 602; http://dx.doi.org/10.1084/jem.20042167; PMID: 15883172
  • Dols A, Smith JW 2nd, Meijer SL, Fox BA, Hu HM, Walker E, Rosenheim S, Moudgil T, Doran T, Wood W, et al. 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 - 23; http://dx.doi.org/10.1089/104303403322124828; PMID: 12885350
  • Jiang XP, Yang DC, Elliott RL, Head JF. Vaccination with a mixed vaccine of autogenous and allogeneic breast cancer cells and tumor associated antigens CA15-3, CEA and CA125--results in immune and clinical responses in breast cancer patients. Cancer Biother Radiopharm 2000; 15:495 - 505; http://dx.doi.org/10.1089/cbr.2000.15.495; PMID: 11155821
  • Elliott RL, Head JF. Adjuvant breast cancer vaccine improves disease specific survival of breast cancer patients with depressed lymphocyte immunity. Surg Oncol 2013; 22:172 - 7; http://dx.doi.org/10.1016/j.suronc.2013.05.003; PMID: 23791552
  • 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 - 7; http://dx.doi.org/10.1200/JCO.2004.10.197; PMID: 15254047
  • Nemunaitis J, Dillman RO, Schwarzenberger PO, Senzer N, Cunningham C, Cutler J, Tong A, Kumar P, Pappen B, Hamilton C, et al. Phase II study of belagenpumatucel-L, a transforming growth factor beta-2 antisense gene-modified allogeneic tumor cell vaccine in non-small-cell lung cancer. J Clin Oncol 2006; 24:4721 - 30; http://dx.doi.org/10.1200/JCO.2005.05.5335; PMID: 16966690
  • Nemunaitis J, Nemunaitis M, Senzer N, Snitz P, Bedell C, Kumar P, Pappen B, Maples PB, Shawler D, Fakhrai H. Phase II trial of Belagenpumatucel-L, a TGF-beta2 antisense gene modified allogeneic tumor vaccine in advanced non small cell lung cancer (NSCLC) patients. Cancer Gene Ther 2009; 16:620 - 4; http://dx.doi.org/10.1038/cgt.2009.15; PMID: 19287371
  • Raez LE, Cassileth PA, Schlesselman JJ, Padmanabhan S, Fisher EZ, Baldie PA, Sridhar K, Podack ER. Induction of CD8 T-cell-Ifn-gamma response and positive clinical outcome after immunization with gene-modified allogeneic tumor cells in advanced non-small-cell lung carcinoma. Cancer Gene Ther 2003; 10:850 - 8; http://dx.doi.org/10.1038/sj.cgt.7700641; PMID: 14605671
  • Raez L, Walker G, Baldie P, Fisher E, Gomez J, Tolba K, Santos E, Podack E. CD8 T cell response in a phase I study of therapeutic vaccination of advanced NSCLC with allogeneic tumor cells secreting endoplasmic reticulum-chaperone gp96-Ig-peptide complexes. Advances in Lung Cancer 2013; 2:9 - 18; http://dx.doi.org/10.4236/alc.2013.21002
  • Hus I, Roliński J, Tabarkiewicz J, Wojas K, Bojarska-Junak A, Greiner J, Giannopoulos K, Dmoszyńska A, Schmitt M. Allogeneic dendritic cells pulsed with tumor lysates or apoptotic bodies as immunotherapy for patients with early-stage B-cell chronic lymphocytic leukemia. Leukemia 2005; 19:1621 - 7; http://dx.doi.org/10.1038/sj.leu.2403860; PMID: 15990861
  • Smith BD, Kasamon YL, Kowalski J, Gocke C, Murphy K, Miller CB, Garrett-Mayer E, Tsai HL, Qin L, Chia C, et al. K562/GM-CSF immunotherapy reduces tumor burden in chronic myeloid leukemia patients with residual disease on imatinib mesylate. Clin Cancer Res 2010; 16:338 - 47; http://dx.doi.org/10.1158/1078-0432.CCR-09-2046; PMID: 20048335
  • Morton DL, Hsueh EC, Essner R, Foshag LJ, O’Day SJ, Bilchik A, Gupta RK, Hoon DS, Ravindranath M, Nizze JA, et al. Prolonged survival of patients receiving active immunotherapy with Canvaxin therapeutic polyvalent vaccine after complete resection of melanoma metastatic to regional lymph nodes. Ann Surg 2002; 236:438 - 48, discussion 448-9; http://dx.doi.org/10.1097/00000658-200210000-00006; PMID: 12368672
  • Sondak VK, Sosman JA. Results of clinical trials with an allogenic melanoma tumor cell lysate vaccine: Melacine. Semin Cancer Biol 2003; 13:409 - 15; http://dx.doi.org/10.1016/j.semcancer.2003.09.004; PMID: 15001159
  • Mitchell MS, Kan-Mitchell J, Kempf RA, Harel W, Shau HY, Lind S. Active specific immunotherapy for melanoma: phase I trial of allogeneic lysates and a novel adjuvant. Cancer Res 1988; 48:5883 - 93; PMID: 3262416
  • Vaishampayan U, Abrams J, Darrah D, Jones V, Mitchell MS. Active immunotherapy of metastatic melanoma with allogeneic melanoma lysates and interferon alpha. Clin Cancer Res 2002; 8:3696 - 701; PMID: 12473578
  • Sosman JA, Unger JM, Liu PY, Flaherty LE, Park MS, Kempf RA, Thompson JA, Terasaki PI, Sondak VK, Southwest Oncology Group. Adjuvant immunotherapy of resected, intermediate-thickness, node-negative melanoma with an allogeneic tumor vaccine: impact of HLA class I antigen expression on outcome. J Clin Oncol 2002; 20:2067 - 75; http://dx.doi.org/10.1200/JCO.2002.08.072; PMID: 11956267
  • Wang F, Bade E, Kuniyoshi C, Spears L, Jeffery G, Marty V, Groshen S, Weber J. Phase I trial of a MART-1 peptide vaccine with incomplete Freund’s adjuvant for resected high-risk melanoma. Clin Cancer Res 1999; 5:2756 - 65; PMID: 10537339
  • von Euw EM, Barrio MM, Furman D, Bianchini M, Levy EM, Yee C, Li Y, Wainstok R, Mordoh J. Monocyte-derived dendritic cells loaded with a mixture of apoptotic/necrotic melanoma cells efficiently cross-present gp100 and MART-1 antigens to specific CD8(+) T lymphocytes. J Transl Med 2007; 5:19; http://dx.doi.org/10.1186/1479-5876-5-19; PMID: 17448240
  • Palucka AK, Ueno H, Connolly J, Kerneis-Norvell F, Blanck J-P, Johnston DA, Fay J, Banchereau J. Dendritic cells loaded with killed allogeneic melanoma cells can induce objective clinical responses and MART-1 specific CD8+ T-cell immunity. J Immunother 2006; 29:545 - 57; http://dx.doi.org/10.1097/01.cji.0000211309.90621.8b; PMID: 16971810
  • Michael A, Ball G, Quatan N, Wushishi F, Russell N, Whelan J, Chakraborty P, Leader D, Whelan M, Pandha H. Delayed disease progression after allogeneic cell vaccination in hormone-resistant prostate cancer and correlation with immunologic variables. Clin Cancer Res 2005; 11:4469 - 78; http://dx.doi.org/10.1158/1078-0432.CCR-04-2337; PMID: 15958632
  • Eaton JD, Perry MJA, Nicholson S, Guckian M, Russell N, Whelan M, Kirby RS. Allogeneic whole-cell vaccine: a phase I/II study in men with hormone-refractory prostate cancer. BJU Int 2002; 89:19 - 26; http://dx.doi.org/10.1046/j.1464-410X.2002.02572.x; PMID: 11849155
  • Hrouda D, Souberbielle BE, Kayaga J, Corbishley CM, Kirby RS, Dalgleish AG. Mycobacterium vaccae (SRL172): a potential immunological adjuvant evaluated in rat prostate cancer. Br J Urol 1998; 82:870 - 6; http://dx.doi.org/10.1046/j.1464-410X.1998.00881.x; PMID: 9883227
  • Small EJ, Sacks N, Nemunaitis J, Urba WJ, Dula E, Centeno AS, Nelson WG, Ando D, Howard C, Borellini F, et al. Granulocyte macrophage colony-stimulating factor--secreting allogeneic cellular immunotherapy for hormone-refractory prostate cancer. Clin Cancer Res 2007; 13:3883 - 91; http://dx.doi.org/10.1158/1078-0432.CCR-06-2937; PMID: 17606721
  • Kaighn ME, Narayan KS, Ohnuki Y, Lechner JF, Jones LW. Establishment and characterization of a human prostatic carcinoma cell line (PC-3). Invest Urol 1979; 17:16 - 23; PMID: 447482
  • Montgomery BT, Young CYF, Bilhartz DL, Andrews PE, Prescott JL, Thompson NF, Tindall DJ. Hormonal regulation of prostate-specific antigen (PSA) glycoprotein in the human prostatic adenocarcinoma cell line, LNCaP. Prostate 1992; 21:63 - 73; http://dx.doi.org/10.1002/pros.2990210107; PMID: 1379363
  • Eager R, Nemunaitis J. GM-CSF gene-transduced tumor vaccines. Mol Ther 2005; 12:18 - 27; http://dx.doi.org/10.1016/j.ymthe.2005.02.012; PMID: 15963916
  • Higano CS, Corman JM, Smith DC, Centeno AS, Steidle CP, Gittleman M, Simons JW, Sacks N, Aimi J, Small EJ. Phase 1/2 dose-escalation study of a GM-CSF-secreting, allogeneic, cellular immunotherapy for metastatic hormone-refractory prostate cancer. Cancer 2008; 113:975 - 84; http://dx.doi.org/10.1002/cncr.23669; PMID: 18646045
  • Corman JM, Small EJ, Smith DC, Centeno AS, Gittelman M, Steidle CP, et al. Immunotherapy with GVAX((R)) vaccine for prostate cancer improves predicted survival in metastatic hormone refractory prostate cancer: Results from two phase 2 studies.
  • Lassi K, Dawson NA. Update on castrate-resistant prostate cancer: 2010. Curr Opin Oncol 2010; 22:263 - 7; http://dx.doi.org/10.1097/CCO.0b013e3283380939; PMID: 20177381
  • Chambers CA, Kuhns MS, Egen JG, Allison JP. CTLA-4-mediated inhibition in regulation of T cell responses: Mechanisms and manipulation in tumor immunotherapy. Annual Review of Immunology: Annual Reviews {a}, 4139 El Camino Way, Palo Alto, CA, 94303-0139, USA, 2001:565-94.
  • Nguyen M, Koprivnikar K, Guang H-T, Jooss K, Harding T. Identification of antibody responses induced in patients with metastatic hormone-refractory prostate cancer (HRPC) treated with a GM-CSF-transduced allogeneic prostate cancer immunotherapy (GVAX) and ipilimumab. J Immunother 2007; 30:900
  • Pohla H, Brill T, Kuebler H, Buchner A, van Randenborgh H, Paul R, et al. Allogeneic Retrovirally Transduced, IL-2 and IFN-gamma Secreting Tumor Cell Vaccine in Patients With Hormone-Refractory Prostate Cancer (HRPC)-A Phase I/II Trial. J Immunother 2008; 31:947 - 8
  • Dodson LF, Hawkins WG, Goedegebuure P. Potential targets for pancreatic cancer immunotherapeutics. Immunotherapy 2011; 3:517 - 37; http://dx.doi.org/10.2217/imt.11.10; PMID: 21463193
  • Laheru D, Biedrzycki B, Thomas AM, Jaffee EM. Development of a cytokine-modified allogeneic whole cell pancreatic cancer vaccine. In: Su GH, ed. Methods in Molecular Medicine, 2005:299-327.
  • Middleton G, Ghaneh P, Costello E, Greenhalf W, Neoptolemos JP. New treatment options for advanced pancreatic cancer. Expert Rev Gastroenterol Hepatol 2008; 2:673 - 96; http://dx.doi.org/10.1586/17474124.2.5.673; PMID: 19072345
  • Galili U, LaTemple DC. Natural anti-Gal antibody as a universal augmenter of autologous tumor vaccine immunogenicity. Immunol Today 1997; 18:281 - 5; http://dx.doi.org/10.1016/S0167-5699(97)80024-2; PMID: 9190114
  • Macher BA, Galili U. The Galalpha1,3Galbeta1,4GlcNAc-R (alpha-Gal) epitope: a carbohydrate of unique evolution and clinical relevance. Biochim Biophys Acta 2008; 1780:75 - 88; http://dx.doi.org/10.1016/j.bbagen.2007.11.003; PMID: 18047841
  • LaTemple DC, Henion TR, Anaraki F, Galili U. Synthesis of alpha-galactosyl epitopes by recombinant alpha1,3galactosyl transferase for opsonization of human tumor cell vaccines by anti-galactose. Cancer Res 1996; 56:3069 - 74; PMID: 8674064
  • Hardacre JM, Mulcahy M, Small W, Talamonti M, Obel J, Krishnamurthi S, Rocha-Lima CS, Safran H, Lenz HJ, Chiorean EG. Addition of algenpantucel-L immunotherapy to standard adjuvant therapy for pancreatic cancer: a phase 2 study. J Gastrointest Surg 2013; 17:94 - 100, discussion 100-1; http://dx.doi.org/10.1007/s11605-012-2064-6; PMID: 23229886
  • Jaffee EM, Hruban RH, Biedrzycki B, Laheru D, Schepers K, Sauter PR, Goemann M, Coleman J, Grochow L, Donehower RC, et al. Novel allogeneic granulocyte-macrophage colony-stimulating factor-secreting tumor vaccine for pancreatic cancer: a phase I trial of safety and immune activation. J Clin Oncol 2001; 19:145 - 56; PMID: 11134207
  • Laheru D, Lutz E, Burke J, Biedrzycki B, Solt S, Onners B, Tartakovsky I, Nemunaitis J, Le D, Sugar E, et al. Allogeneic granulocyte macrophage colony-stimulating factor-secreting tumor immunotherapy alone or in sequence with cyclophosphamide for metastatic pancreatic cancer: a pilot study of safety, feasibility, and immune activation. Clin Cancer Res 2008; 14:1455 - 63; http://dx.doi.org/10.1158/1078-0432.CCR-07-0371; PMID: 18316569
  • Lutz E, Yeo CJ, Lillemoe KD, Biedrzycki B, Kobrin B, Herman J, Sugar E, Piantadosi S, Cameron JL, Solt S, et al. A lethally irradiated allogeneic granulocyte-macrophage colony stimulating factor-secreting tumor vaccine for pancreatic adenocarcinoma. A Phase II trial of safety, efficacy, and immune activation. Ann Surg 2011; 253:328 - 35; http://dx.doi.org/10.1097/SLA.0b013e3181fd271c; PMID: 21217520
  • Habal N, Gupta RK, Bilchik AJ, Yee R, Leopoldo Z, Ye W, Elashoff RM, Morton DL. CancerVax, an allogeneic tumor cell vaccine, induces specific humoral and cellular immune responses in advanced colon cancer. Ann Surg Oncol 2001; 8:389 - 401; http://dx.doi.org/10.1007/s10434-001-0389-6; PMID: 11407512
  • Westermann J, Flörcken A, Willimsky G, van Lessen A, Kopp J, Takvorian A, Jöhrens K, Lukowsky A, Schönemann C, Sawitzki B, et al. Allogeneic gene-modified tumor cells (RCC-26/IL-7/CD80) as a vaccine in patients with metastatic renal cell cancer: a clinical phase-I study. Gene Ther 2011; 18:354 - 63; http://dx.doi.org/10.1038/gt.2010.143; PMID: 21068778
  • Emens LA, Machiels JP, Reilly RT, Jaffee EM. Chemotherapy: friend or foe to cancer vaccines?. Curr Opin Mol Ther 2001; 3:77 - 84; PMID: 11249735
  • Glöckner S, Buurman H, Kleeberger W, Lehmann U, Kreipe H. Marked intratumoral heterogeneity of c-myc and cyclinD1 but not of c-erbB2 amplification in breast cancer. Lab Invest 2002; 82:1419 - 26; http://dx.doi.org/10.1097/01.LAB.0000032371.16521.40; PMID: 12379776
  • Yap TA, Gerlinger M, Futreal PA, Pusztai L, Swanton C. Intratumor heterogeneity: seeing the wood for the trees. Sci Transl Med 2012; 4:27ps10; http://dx.doi.org/10.1126/scitranslmed.3003854; PMID: 22461637
  • Curtis C, Shah SP, Chin SF, Turashvili G, Rueda OM, Dunning MJ, Speed D, Lynch AG, Samarajiwa S, Yuan Y, et al, METABRIC Group. The genomic and transcriptomic architecture of 2,000 breast tumours reveals novel subgroups. Nature 2012; 486:346 - 52; PMID: 22522925
  • Rakha EA, Boyce RWG, Abd El-Rehim D, Kurien T, Green AR, Paish EC, Robertson JF, Ellis IO. Expression of mucins (MUC1, MUC2, MUC3, MUC4, MUC5AC and MUC6) and their prognostic significance in human breast cancer. Mod Pathol 2005; 18:1295 - 304; http://dx.doi.org/10.1038/modpathol.3800445; PMID: 15976813
  • Springer GF. Immunoreactive T and Tn epitopes in cancer diagnosis, prognosis, and immunotherapy. J Mol Med (Berl) 1997; 75:594 - 602; http://dx.doi.org/10.1007/s001090050144; PMID: 9297627
  • Brockhausen I. Mucin-type O-glycans in human colon and breast cancer: glycodynamics and functions. EMBO Rep 2006; 7:599 - 604; http://dx.doi.org/10.1038/sj.embor.7400705; PMID: 16741504
  • Fitzgibbons PL, Page DL, Weaver D, Thor AD, Allred DC, Clark GM, Ruby SG, O’Malley F, Simpson JF, Connolly JL, et al. Prognostic factors in breast cancer. College of American Pathologists Consensus Statement 1999. Arch Pathol Lab Med 2000; 124:966 - 78; PMID: 10888772
  • Novellino L, Castelli C, Parmiani G. A listing of human tumor antigens recognized by T cells: March 2004 update. Cancer Immunol Immunother 2005; 54:187 - 207; http://dx.doi.org/10.1007/s00262-004-0560-6; PMID: 15309328
  • Greiner J, Schmitt M, Li L, Giannopoulos K, Bosch K, Schmitt A, Dohner K, Schlenk RF, Pollack JR, Dohner H, et al. Expression of tumor-associated antigens in acute myeloid leukemia: Implications for specific immunotherapeutic approaches. Blood 2006; 108:4109 - 17; http://dx.doi.org/10.1182/blood-2006-01-023127; PMID: 16931630
  • Lee PP, Yee C, Savage PA, Fong L, Brockstedt D, Weber JS, Johnson D, Swetter S, Thompson J, Greenberg PD, et al. Characterization of circulating T cells specific for tumor-associated antigens in melanoma patients. Nat Med 1999; 5:677 - 85; http://dx.doi.org/10.1038/9525; PMID: 10371507
  • Hassan R, Ho M. Mesothelin targeted cancer immunotherapy. Eur J Cancer 2008; 44:46 - 53; http://dx.doi.org/10.1016/j.ejca.2007.08.028; PMID: 17945478
  • Ghaneh P, Costello E, Neoptolemos JP. Biology and management of pancreatic cancer. Gut 2007; 56:1134 - 52; PMID: 17625148
  • Emens LA, Armstrong D, Biedrzycki B, Davidson N, Davis-Sproul J, Fetting J, Jaffee E, Onners B, Piantadosi S, Reilly RT, et al. A phase I vaccine safety and chemotherapy dose-finding trial of an allogeneic GM-CSF-secreting breast cancer vaccine given in a specifically timed sequence with immunomodulatory doses of cyclophosphamide and doxorubicin. Hum Gene Ther 2004; 15:313 - 37; http://dx.doi.org/10.1089/104303404322886165; PMID: 15018740
  • Emens LA, Asquith JM, Leatherman JM, Kobrin BJ, Petrik S, Laiko M, Levi J, Daphtary MM, Biedrzycki B, Wolff AC, et al. Timed sequential treatment with cyclophosphamide, doxorubicin, and an allogeneic granulocyte-macrophage colony-stimulating factor-secreting breast tumor vaccine: a chemotherapy dose-ranging factorial study of safety and immune activation. J Clin Oncol 2009; 27:5911 - 8; http://dx.doi.org/10.1200/JCO.2009.23.3494; PMID: 19805669
  • Yannelli JR, Sturgill J, Foody T, Hirschowitz E. The large scale generation of dendritic cells for the immunization of patients with non-small cell lung cancer (NSCLC). Lung Cancer 2005; 47:337 - 50; http://dx.doi.org/10.1016/j.lungcan.2004.08.008; PMID: 15713517
  • Hirschowitz EA, Foody T, Hidalgo GE, Yannelli JR. Immunization of NSCLC patients with antigen-pulsed immature autologous dendritic cells. Lung Cancer 2007; 57:365 - 72; http://dx.doi.org/10.1016/j.lungcan.2007.04.002; PMID: 17509725
  • Nemunaitis J, Jahan T, Ross H, Sterman D, Richards D, Fox B, Jablons D, Aimi J, Lin A, Hege K. Phase 1/2 trial of autologous tumor mixed with an allogeneic GVAX vaccine in advanced-stage non-small-cell lung cancer. Cancer Gene Ther 2006; 13:555 - 62; http://dx.doi.org/10.1038/sj.cgt.7700922; PMID: 16410826
  • von Euw EM, Barrio MM, Furman D, Levy EM, Bianchini M, Peguillet I, Lantz O, Vellice A, Kohan A, Chacón M, et al. A phase I clinical study of vaccination of melanoma patients with dendritic cells loaded with allogeneic apoptotic/necrotic melanoma cells. Analysis of toxicity and immune response to the vaccine and of IL-10 -1082 promoter genotype as predictor of disease progression. J Transl Med 2008; 6:6; http://dx.doi.org/10.1186/1479-5876-6-6; PMID: 18221542
  • Mordoh J, Kairiyama C, Bover L, Solarolo E. Allogeneic cells vaccine increases disease-free survival in stage III melanoma patients. A non randomized phase II study. Medicina (B Aires) 1997; 57:421 - 7; PMID: 9674264
  • Barrio MM, de Motta PT, Kaplan J, von Euw EM, Bravo AI, Chacón RD, Mordoh J. A phase I study of an allogeneic cell vaccine (VACCIMEL) with GM-CSF in melanoma patients. J Immunother 2006; 29:444 - 54; http://dx.doi.org/10.1097/01.cji.0000208258.79005.5f; PMID: 16799340
  • Simons JW, Carducci MA, Mikhak B, Lim M, Biedrzycki B, Borellini F, Clift SM, Hege KM, Ando DG, Piantadosi S, et al. Phase I/II trial of an allogeneic cellular immunotherapy in hormone-naïve prostate cancer. Clin Cancer Res 2006; 12:3394 - 401; http://dx.doi.org/10.1158/1078-0432.CCR-06-0145; PMID: 16740763
  • van den Eertwegh AJM, Versluis J, van den Berg HP, Santegoets SJAM, van Moorselaar RJA, van der Sluis TM, Gall HE, Harding TC, Jooss K, Lowy I, et al. Combined immunotherapy with granulocyte-macrophage colony-stimulating factor-transduced allogeneic prostate cancer cells and ipilimumab in patients with metastatic castration-resistant prostate cancer: a phase 1 dose-escalation trial. Lancet Oncol 2012; 13:509 - 17; http://dx.doi.org/10.1016/S1470-2045(12)70007-4; PMID: 22326922
  • Brill TH, Kübler HR, von Randenborgh H, Fend F, Pohla H, Breul J, Hartung R, Paul R, Schendel DJ, Gansbacher B. Allogeneic retrovirally transduced, IL-2- and IFN-gamma-secreting cancer cell vaccine in patients with hormone refractory prostate cancer--a phase I clinical trial. J Gene Med 2007; 9:547 - 60; http://dx.doi.org/10.1002/jgm.1051; PMID: 17514769
  • Brill TH, Kübler HR, Pohla H, Buchner A, Fend F, Schuster T, van Randenborgh H, Paul R, Kummer T, Plank C, et al. Therapeutic vaccination with an interleukin-2-interferon-gamma-secreting allogeneic tumor vaccine in patients with progressive castration-resistant prostate cancer: a phase I/II trial. Hum Gene Ther 2009; 20:1641 - 51; http://dx.doi.org/10.1089/hum.2009.101; PMID: 19671000

Reprints and Corporate Permissions

Please note: Selecting permissions does not provide access to the full text of the article, please see our help page How do I view content?

To request a reprint or corporate permissions for this article, please click on the relevant link below:

Academic Permissions

Please note: Selecting permissions does not provide access to the full text of the article, please see our help page How do I view content?

Obtain permissions instantly via Rightslink by clicking on the button below:

If you are unable to obtain permissions via Rightslink, please complete and submit this Permissions form. For more information, please visit our Permissions help page.