Skip to main content

A Phase I vaccine trial using dendritic cells pulsed with autologous oxidized lysate for recurrent ovarian cancer

Abstract

Purpose

Ovarian cancer, like most solid tumors, is in dire need of effective therapies. The significance of this trial lies in its promise to spearhead the development of combination immunotherapy and to introduce novel approaches to therapeutic immunomodulation, which could enable otherwise ineffective vaccines to achieve clinical efficacy.

Rationale

Tumor-infiltrating T cells have been associated with improved outcome in ovarian cancer, suggesting that activation of antitumor immunity will improve survival. However, molecularly defined vaccines have been generally disappointing. Cancer vaccines elicit a modest frequency of low-to-moderate avidity tumor-specific T-cells, but powerful tumor barriers dampen the engraftment, expansion and function of these effector T-cells in the tumor, thus preventing them from reaching their full therapeutic potential. Our work has identified two important barriers in the tumor microenvironment: the blood-tumor barrier, which prevents homing of effector T cells, and T regulatory cells, which inactivate effector T cells. We hypothesize that cancer vaccine therapy will benefit from combinations that attenuate these two barrier mechanisms.

Design

We propose a three-cohort sequential study to investigate a combinatorial approach of a new dendritic cell (DC) vaccine pulsed with autologous whole tumor oxidized lysate, in combination with antiangiogenesis therapy (bevacizumab) and metronomic cyclophosphamide, which impacts Treg cells.

Innovation

This study uses a novel autologous tumor vaccine developed with 4-day DCs pulsed with oxidized lysate to elicit antitumor response. Furthermore, the combination of bevacizumab with a whole tumor antigen vaccine has not been tested in the clinic. Finally the combination of bevacizumab and metronomic cyclophosphamide in immunotherapy is novel.

Background and rationale

Ovarian cancer cells are antigenic and express a multitude of known tumor-associated antigens (TAAs) including Her-2/neu [13], p53 [4], NY-ESO-1 [5, 6], cdr2 [7], hTERT [8, 9], mesothelin [10], survivin [11, 12] SP-17, WT1 [1319] etc. Clinical data now clearly indicate that the immune system affects the outcome of patients with epithelial ovarian cancer (EOC). We and others have shown that the presence of intraepithelial tumor-infiltrating lymphocytes correlates with improved progression-free and overall survival [2028]. TILs isolated from ovarian cancers are oligoclonal [29, 30], recognize autologous tumor and known TAAs in vitro [14, 3134], and exhibit tumor-specific cytolytic activity ex vivo [35, 36]. Tumor-specific T-cell precursors can also be detected in the blood of patients with advanced ovarian carcinoma [37]. These observations suggest that activation of antitumor immunity could be feasible and could produce clinical results. To date, immunotherapy investigations have yielded limited but encouraging results in EOC. For example, weekly intraperitoneal IL-2 infusion produced a ~17% complete pathologic response rate in ovarian cancer [38, 39], while anecdotal objective responses have been reported with CTLA-4 antibody [40, 41]; adoptive transfer of TILs [23, 24]; or vaccines using NY-ESO-1 peptide [42], virus-modified autologous tumor cells [43] or DCs pulsed with whole autologous tumor lysate [44].

Whole tumor cancer vaccines

Therapeutic cancer vaccines have the potential to break immune tolerance and induce long-term immune response against cancer cells. However, molecularly defined vaccines directed towards known TAAs have either failed to produce clinical responses or have yielded transient responses in ovarian cancer patients to date, as none of the above antigens, except for NY-ESO-1, have been proven to be bona fide rejection antigens in the clinic [42, 45, 46]. A reasonable alternative may be whole tumor vaccines [4749]. The advantages of these were reviewed recently [50]. Tumor cells express a whole array of antigens, most of which remain uncharacterized in EOC. Vaccination with whole tumor antigen potentially draws on this rich source of antigens, comprising epitopes for both CD8+ cytotoxic T-cells (CTLs) as well as CD4+ T helper (Th) cells, a possibly necessary condition to ensure tumor homing of low affinity CD8+ cells [5153]. Whole tumor vaccines could also greatly diminish the chance of tumor escape compared to single epitope vaccines. Finally, recent deep sequencing results from over 300 advanced EOC specimens show that ovarian tumors carry an average of 61 somatic non-synonymous mutations, most of which were private [54]. Some of these mutations could potentially give rise to neo-antigens that could stimulate effective and long-lasting anti-tumor responses. Interestingly, a meta-analysis of 173 published peer-reviewed immunotherapy trials found that 8.1% of patients vaccinated with whole tumor antigen (n=1,733) experienced objective clinical responses, compared with 3.6% of patients vaccinated with defined tumor antigens (n= 1,711; P < 0.0001) [55]. Although, studies have shown that whole tumor lysates can be poorly immunogenic and can suppress DC differentiation and maturation [5658], some approaches to lysate preparation can increase immunogenicity of whole tumor lysates [5961]. In this study, we exploit oxidation during the preparation of tumor lysate, which appears to promote immunogenicity [62].

A novel approach to tumor cell lysate preparation

A widely used and straightforward method of whole tumor cell preparation already used in clinical trials is necrotic whole tumor cell lysate. The efficacy of the necrotic cell lysate can be further enhanced by oxidative modification using hypochlorous acid (HOCl) treatment [59]. It has been demonstrated that proteins oxidized by HOCl are more readily taken up and processed by antigen presenting cells (APCs) and lead to enhanced priming of autologous tumor-specific CD4+ and CD8+ T-cell responses in vitro [6366]. The use of HOCl to potentiate the immunogenicity of whole ovarian tumor cells has been evaluated using SKOV3 ovarian cancer cells [67, 68]. The improvement in antigen immunogenicity is explained by three possible mechanisms. First, HOCl can quantitatively deaminate serine and convert its side chain into an aldehyde, leading to significant improvement in immunogenicity [6971]. Second, oxidation of protein antigens might allow protein unfolding and exposure of cryptic immunogenic peptides to specific T-cells [72]. Third, scavenger receptors such as the lectin-like oxidized low-density lipoprotein receptor-1 (LOX-1) might be involved in the uptake of HOCl-oxidized tumor cells [7375], leading to DC activation and efficient presentation of MHC-I as well as MHC-II restricted peptides [59, 76]. In preclinical evaluation, this tumor lysate preparation proved to be more immunogenic than the standard UV treated whole tumor lysate [50]. This will be the first study utilizing DCs pulsed with oxidized whole tumor lysate.

A new dendritic cell vaccine platform

DCs loaded with whole tumor lysate have been investigated in several clinical trials for their ability to induce anti-tumor T-cell responses [44, 7780]. Beneficial anti-tumor responses have been observed in some patients, illustrating the potential of this approach. DCs can be classified into different subsets, depending on their lineage and receptor expression pattern. Their distinct biology can be exploited for different therapeutic strategies. The most widely used DCs in clinical trials to date are myeloid DCs differentiated from peripheral blood monocytes. In most trials, “classic” DCs are fully differentiated over seven days in the presence of recombinant granulocyte-macrophage colony stimulating factor (GM-CSF) and interleukin 4 (IL-4) [8183]. These DCs exhibit high phagocytic and antigen-processing capability. Upon maturation with an appropriate stimulus, Day-7 DCs up regulate costimulatory surface molecules such as CD80, CD86, CD40, and lymph node-homing receptors such as CCR7, and can efficiently prime naïve T-cells [8486].

We developed a faster, four-day protocol for DC preparation, using GM-CSF, IL-4 and serum-free AIM-V media that is suitable for clinical use. We showed that Day-4 DCs generated with this protocol are similar to “classic” Day-7 DCs, in terms of phenotype and phagocytic capability, and have a higher capacity than Day-7 DCs to produce IL-12p70 following LPS and IFN-γ stimulation. In addition, these Day-4 DCs were highly immunogenic, and efficiently primed ovarian tumor-specific T-cells in vitro in peripheral blood lymphocytes from healthy volunteers and ovarian cancer patients [87].

Enhancement of immune therapy by antiangiogenic therapy

It has been shown that vascular endothelial growth factor (VEGF) suppresses tumor antigen presentation through blockade of myeloid DC differentiation and maturation, leading to tumor immune tolerance [8895], a process primarily mediated by VEGF receptor-1 (VEGFR-1) [96]. VEGF also up regulates programmed death ligand 1 (PD-L1 or B7-H1) in myeloid DCs, which is associated with T-cell suppression and exhaustion [97]. VEGF blockade restores DC function and enhances immunotherapy [12, 98101]. Anti-VEGF strategies reduce the number of CD4+CD25+ T regulatory (Treg) cells when administered in combination with a GM-CSF secreting tumor vaccine, resulting in increased CTL induction and improved vaccine efficacy [102, 103]. Similar effects were observed in human subjects treated with VEGF-neutralizing antibody therapy [98]. A single-arm clinical trial of vaccine and bevacizumab for prostate cancer has shown that the combination is associated with a high rate of immune response induction [104].

Work by us and others have demonstrated that angiogenesis mechanisms also impair the effector arm of antitumor immunity by blocking homing of effector T-cells into tumors. This is in part mediated through the endothelin B receptor (ETBR), which is up regulated in tumor endothelium and deregulates endothelial ICAM-1 expression and T-cell adhesion. This blood-tumor endothelial barrier can be disrupted by ETBR antagonists, resulting in dramatic increase of T-cell homing in tumors and significant efficacy of otherwise ineffective vaccine therapy in the mouse, thus calling for human experimentation [105, 106]. Importantly, ETBR is upregulated by VEGF. In vitro, ET-1 signaling through ETAR in cancer cell lines leads to de novo production of VEGF, a process regulated by HIF-1-alpha [107] utilizing initial PGE2 production [108] as an intermediary before secondary VEGF production [107, 109112]. We confirmed that VEGF blockade enhances T-cell homing to tumors in ID8-VEGF tumors, a murine syngeneic model of ovarian cancer overexpressing VEGF [113]. Following vaccination with UV-irradiated tumor cells, in spite of a tangible frequency of antitumor T-cells in the spleen, no CD8+ TILs were detected in ID8-VEGF tumors. In agreement with others [114], treatment with the VEGFR-2 tyrosine kinase inhibitor SU5416 [115] produced a dramatic influx of CD8+ TILs in ID8-VEGF tumors, while DMSO vehicle had no effect on TILs (unpublished data). Normalization of tumor vasculature through disruption of the VEGF/VEGFR-2 axis was also shown to increase extravasation of adoptively transferred T-cells into the tumor and improve adoptive cell transfer immunotherapy in a murine cancer model [116]. Combining VEGFR-2 antibody DC101 with Her-2-specific vaccination in a mouse model of Her-2/neu-induced breast cancer, it was demonstrated that this combination treatment accelerated tumor regression augmenting the lytic activity of CD8+ cytotoxic T-cells [114]. Thus, VEGF blockade not only blocks tumor angiogenesis, but may also increase the efficacy of tumor vaccines by enhancing DC function and by increasing T-cell homing to tumors.

Using metronomic chemotherapy to enhance immune response

Although, the traditional view has been that chemotherapy may neutralize antitumor immune response generated through vaccine therapy, emergent data indicate that chemotherapy can be combined safely with immunotherapy with possibly additive or synergistic effects that are dose and schedule dependent. The most extensively investigated drug to enhance vaccine potency is cyclophosphamide, a drug previously used in ovarian cancer as standard of care in combination with cisplatin. One of the first observation was made by Berd and colleagues who used a regimen of low-dose cyclophosphamide (300 mg/m2 i.v.), given three days prior to vaccination with autologous melanoma cells admixed with Bacillus Calmette-Guerin (BCG), to treat patients with melanoma [117]. They reported that cyclophosphamide plus vaccine treatment resulted in a progressive depletion of circulating CD4+ suppressor T-cells.

In mouse models, North presented evidence that intravenous cyclophosphamide enhanced tumor immunotherapy by elimination of CD8+ tumor suppressor cells [118], and that the effect was independent of direct tumor cytotoxic effects [119]. Machiels and colleagues evaluated the combination of cyclophosphamide with (GM-CSF)–secreting whole-cell vaccines in the HER-2/neu mouse model of mammary cancers [120]. They found that when cyclophosphamide was given at a dose range between 50 and 150 mg/kg 1 day prior to vaccine, the combination controlled tumors more effectively than either agent alone. Subsequent work showed that cyclophosphamide depletes Treg cells [121, 122] and it impairs their function for nearly 10 days post treatment.

Cyclophosphamide has been used also in humans to augment cancer immunotherapy [123, 124]. In patients with advanced colorectal carcinoma and melanoma, cyclophosphamide was shown to increase the response to an adjuvant KLH vaccine. Jaffee and colleagues have examined carefully the dose-dependent immunomodulatory effects of cyclophosphamide with respect to targeting T regulatory (Treg) cells [125]. Emens et al. conducted a Phase I trial that evaluated allogeneic, Her-2-positive GM-CSF–secreting breast tumor vaccine alone or in sequence with low doses of cyclophosphamide and doxorubicin in metastatic breast cancer patients (n=28), and they found that the dose of 200 mg/m2 of intravenous cyclophosphamide augmented Her-2-specific humoral immunity. However, the immunomodulatory effect of intravenous cyclophosphamide was lost if given at doses above 200 mg/m2[125]. Single-agent intravenous cyclophosphamide doses of 150, 250, and 350 mg/m2 were also evaluated in patients with hepatocellular carcinoma by Greten et al. who reported that the lower doses (150 and 250 mg/m2) induced a decrease the number and the relative frequency of circulating regulatory T-cells, and that the dose of 250 mg/m2 was able to impair the suppressor function of regulatory T-cells. It was also shown in a phase I trial of pancreatic cancer patients that inhibition of regulatory T-cells (when using 250 mg/m2 intravenous cyclophosphamide) resulted in recruitment of high-avidity effector T-cells to tumors, leading to prolonged progression-free survival and overall survival [126].

It has been hypothesized that combining VEGF blockade with low-dose (metronomic) chemotherapy may have positive antiangiogenic or antitumor effects [127]. In a multi-institutional phase II study (NCI-5789), 29 subjects were treated with bevacizumab 10 mg/kg every 14 days and low-dose oral cyclophosphamide 50 mg daily [128]. The response rate was 28% and 6-month progression free survival rate of 57%. A second phase II prospective study investigated the efficacy and safety of intravenous bevacizumab 10 mg/kg every other week plus oral cyclophosphamide 50 mg daily in fifteen heavily pretreated patients with recurrent ovarian cancer [129]. There was significant activity, with a response rate of 53%. Despite being heavily pretreated, no gastrointestinal perforations were noted. In short, combination of bevacizumab and cyclophosphamide is a promising dual antiangiogenic/tumoristatic and immunomodulatory therapy that is available at the present time. The rationale for combining this therapy with vaccine is to prime the tumor microenvironment and create a favorable milieu to achieve greater efficacy and immune response.

Trial design

We propose a phase I, three-cohort, single-center study to establish the safety and proof of concept of Oxidized tumor Cell pulsed DC (OCDC) vaccine administered intranodally, alone (Cohort 1, n=5 subjects) or in combination with either intravenous bevacizumab (Cohort 2, n=10 subjects) or intravenous bevacizumab and intravenous cyclophosphamide (Cohort 3, n=10 subjects) in subjects with recurrent ovarian, fallopian tube or primary peritoneal cancer (Figure 1). Inclusion criteria require patients 18 years or older, diagnosed with advanced stage disease, with ECOG performance status ≤1, and >6 months expected survival.

Figure 1
figure 1

Clinical trial design.

The components of the vaccine in this study include agents, for which safety has been previously demonstrated to be acceptable. During this current study we intend to manufacture a modified whole tumor vaccine using oxidized whole tumor cell lysate derived from autologous tumor harvested at secondary debulking surgery, pulsed onto autologous dendritic cells (Figure 2). The choice of dendritic cell maturation and the choice of tumor cell preparation are based on previously published data [67, 130].

Figure 2
figure 2

Clinical trial schema.

Regimen

Eligible patients will undergo a 10–15 liter apheresis around day −30 to −15 to harvest peripheral blood mononuclear cells (PBMC) for DC manufacturing. Patients will receive OCDC tumor vaccine in combination with other agents in a design that escalates combinatorial complexity. OCDC is prepared at the Cell and Vaccine Production Facility of the University of Pennsylvania. It will be released in sterile syringes containing ~2.5-5 × 106 DC in 0.55 mL sterile saline. Subjects will receive a total dose of approximately 5–10 × 106 DCs administered through two or more intranodal injections into different normal groin nodes; 0.55 cc containing ~2.5-5 × 106 DCs will be injected per node with a 22 Gauge needle.

Subjects in Cohort 1 will receive five doses of 5–10 × 106 dendritic cells (OCDC vaccine) intranodally, while subjects in Cohort 2 will receive the same five doses of vaccine in combination with intravenous bevacizumab (10 mg/kg) given every two weeks on days 0, 14, 28, 42 and 56. Subjects in Cohort 3 will receive the same five doses of vaccine on days 0, 14, 28, 42 and 56, while bevacizumab (10 mg/kg) plus cyclophosphamide (200 mg/m2) will be given the day before each vaccination.

Subjects will be offered to undergo apheresis within two weeks after vaccine #4 or one to three weeks after vaccine #5 to collect vaccine primed PBL for use in a follow-on study of adoptive T-cell therapy. Each treatment cohort will be evaluated separately, to detect any side effects that may be due to the vaccine in combination with other biological agents. Patients in Cohort 2 will be taken off bevacizumab, if they experience severe adverse events (SAEs) at least likely related to bevacizumab, but will retain the option of continuing OCDC vaccination on the study. Subjects in Cohort 3 will be taken off bevacizumab/cyclophosphamide, if they experience severe adverse events (SAEs) at least likely related to bevacizumab or cyclophosphamide, but will retain the option of continuing OCDC vaccination on the study. Subjects will be taken off the study completely if they experience SAEs, which are possibly, probably or definitely related to OCDC. Termination of enrollment for each cohort will be triggered with ≥2 DLTs (i.e., DLT is any Grade 3 or higher allergic, autoimmune or injection site reaction or any Grade 4 hematologic or non-hematologic toxicity expect fever) in the first 5 subjects and ≥3 DLTs at any time. The rules for early stopping for toxicity do not depend on the availability of immune response data; any subject that gets at least one vaccination injection is included. For this study, 25 evaluable subjects will be treated, and we estimate that up to 30 subjects may need to be enrolled, assuming a 20% failure rate for generating the dendritic cell vaccine.

Subjects will be followed daily for the first 5 days (starting at Day 0), and then biweekly until week 8 (End of Study). At screening and 30 days following the fifth vaccine dose (day 86), subjects will undergo immune assessment. Immune monitoring will be performed on blood samples from all participating patients to assess the vaccine induced antitumor immune response and the composition of circulating T-cell subpopulations. This study obtained approval from national health agencies and from the Institutional Review Board of the University of Pennsylvania and is performed in accordance with the Helsinki Declaration, the International Conference of Harmonization Good Clinical Practice guidelines, and local regulatory requirements. Written informed consent will be obtained from each patient.

Objectives

The primary objective of the study is to determine the feasibility and safety of administering OCDC intranodally alone, in combination with intravenous bevacizumab, or in combination with intravenous bevacizumab plus cyclophosphamide in subjects with recurrent ovarian, fallopian tube or primary peritoneal cancer. The secondary objectives of the study are to obtain pilot data on immunogenicity on OCDC administered intranodally in subjects, to assess the effect of the proposed treatments on peripheral blood regulatory T-cells and on the tumor microenvironment, as well as to evaluate clinical responses up to 114 days.

Statistical methods

Toxicity will be graded by NCI Common Toxicity Criteria (NCI-CTC) Version 4.0 and will be tabled by treatment cohort. Immune response will be evaluated by descriptive statistics, scatter plots of pre- and post-vaccine values of the various immune parameters, and relevant fold changes by treatment cohort. Exploratory longitudinal analyses (repeated measures ANOVA or linear mixed effects models) will be used to examine time trends (e.g., decrease in Treg cells), test for differences between baseline and post-vaccine time points within cohorts and discern differences among cohorts. Clinical responses scored by RECIST criteria will be tabulated by treatment cohort.

Innovation

The proposed first-in- human study is innovative in many ways. First, it translates novel concepts of combinatorial immunomodulation of the tumor microenvironment from the laboratory to the clinic. The notion of blocking VEGF in combination with the whole tumor cancer vaccine is novel and has not been tested in the clinic. Preclinical data in solid tumor mouse models show that blocking VEGF enhances de novo T-cell infiltration into the tumor [114] and, when combined with cancer vaccines or adoptive T-cell therapy, VEGF blockade significantly improves their biological and clinical efficacy [104, 116, 131]. Furthermore, although countering Treg cells in combination with vaccine is not novel, the suppression of Treg cells (by low-dose cyclophosphamide) followed by VEGF blockade is novel in the context of immunotherapy. This drug combination could address the possibility that VEGF blockade could trigger tumor hypoxia, which in turn could induce Treg recruitment and immune tolerance via CCL28 chemokine, as we recently reported [132]. Finally, the vaccine platform used in this study is innovative in its own right. Following careful optimization in the lab [62, 87], we are proposing an autologous vaccine with DCs developed from elutriated monocytes cultured for only four days with GM-CSF and IL-4 and pulsed with lysate of HOCl-oxidized tumor cells, which have not been used in the clinic before. DCs will be matured with LPS/IFN-γ and injected intranodally. Collectively, this approach is novel for ovarian cancer, a disease in dire need of new therapies.

Discussion

We propose a new combinatorial therapy approach to mobilize antitumor immunity against ovarian cancer. The strong association between the presence of intraepithelial T-cells along with other biomarkers of immune activation in tumors and improved clinical outcome suggests that mobilization of antitumor immunity should yield clinical benefit in many patients with EOC, a notion preliminarily supported by many published pilot studies. Our group and other groups have however, revealed the existence of numerous and overlapping mechanisms of immune dysfunction in ovarian cancer, which will have to be abated in order to effectively mobilize antitumor immunity. The combinatorial approach proposed herein is a first attempt to utilize readily available therapeutic tools with known clinical and biological behavior to address some of the tumor barriers.

Given the paucity of reliable tumor rejection antigens in EOC (with the exception of NY-ESO-1, which is relatively rare), we propose to use a whole tumor lysate vaccine. Given the easy accessibility of primary but also recurrent tumors in the peritoneal cavity, and the general acceptance of primary or secondary surgical cytoreductive surgery as the standard of care for these patients, autologous tumor lysate is a feasible approach in this population. Autologous tumor lysates provide a convenient and personalized source of multiple tumor antigens, possibly encompassing all the relevant class I and II epitopes against which antitumor immune response can be mounted, including private and mutated epitopes. Incorporation of class II epitopes could be especially important, since the coexistence of tumor-reactive CD4+ cells can enhance tumor engraftment and persistence of low affinity anti-tumor CD8+ cells [53]. The choice of tumor cell preparation is based on previous data demonstrating that oxidative necrosis enhances the immunogenicity of whole tumor cell lysates and is superior to other conventional tumor lysates [133, 134].

There is no strictly defined standard duration of culture to generate human PBMC-derived DCs. To date, most clinical studies have used a 7-day culture with GM-CSF and IL-4. However, data from Czerniecki and colleagues have shown that fully functional antigen-presenting cells (APC) can be rapidly developed from CD14+ PBMC cells in as little as 40 hours [135, 136]. Although, these “rapid DC” are efficient in presenting class I and II peptides, data from our laboratory revealed that at least four days of differentiation with GM-CSF and IL-4 were required for elutriated peripheral blood monocytes to acquire the phenotype and functional properties of cross-presenting APCs capable of processing lysate antigen [87]. These 4-day DCs pulsed with lysate underwent proper maturation into DC1 cells when exposed to bacterial lipopolysaccharide (LPS) combined with IFN-γ, producing high levels of IL-12, but required a concomitant – not sequential – exposure to the two maturing agents [62]. We selected these cells as the proposed vaccine platform, which will be administered intranodally, since intranodal administration of DCs allows administration of a defined quantity of DCs directly to the site of T-cell sensitization. This approach also allows the peak IL-12 secretion to be synchronized with their proximity to T-cells, where IL-12 can exert its full effects during antigen presentation [137]. IL-12 is paramount as dendritic cells that are able to produce high levels of IL-12 can induce long-lived type 1 T-cell responses against tumor-associated antigens more efficiently than standard mature DCs. The benefit of high IL-12 producing DCs was highlighted in several recent papers [138140] demonstrating the importance of IL-12 production with regard to the induction of tumor-specific CTLs in vitro and its ability to predict prolongation of progression-free survival of patients with advanced cancer [141]. Okada et al. showed in a Phase I/II cancer vaccine trial of malignant glioma patients that IL-12 production levels by αDC1 positively correlated with time to progression.

In a murine tumor model, DC pulsed with tumor lysate and injected intranodally resulted in greater sensitization of T-cells and improved anti-tumor responses [142]. In a randomized, Phase I, dose-escalation trial Lambert et al. compared different administration routes (intravenous, intranodal, intradermal) in metastatic melanoma receiving four autologous peptide-pulsed DC vaccinations. The results showed that intranodal administration led to superior T-cell sensitization as measured by de novo target-cell recognition and DTH priming, indicating that intranodal injection may be the preferred route of administration for mature DC vaccines [143].

Adoptive immunotherapy has frequently resulted in tumor rejection in the human, suggesting that a critical number of high-avidity tumor-reactive T-cells are probably required to effectively overcome barriers in the tumor microenvironment. On the contrary, cancer vaccines have most commonly failed to induce overwhelming tumor responses in patients. Preclinical models show that if one can defeat immune barriers in the tumor microenvironment, one can enable low-avidity/low-frequency antitumor immune responses induced by vaccines to become clinically effective. Work from our lab and from other labs shows that among tumor microenvironment barriers preventing the engraftment, expansion and function of antitumor effector T-cells, two can be readily targeted in ovarian cancer: a) angiogenesis driven by VEGF, and b) Treg cells.

Vascular endothelial growth factor is highly expressed and plays an important role in tumor progression of ovarian carcinoma. Positive immunostaining for VEGF was observed in 97% (68 out of 70) of ovarian carcinomas [144] and high VEGF levels correlated with advanced disease stage and poorer survival [144, 145]. VEGF became a fundamental target in anti-angiogenic therapy leading to the development of humanized recombinant monocloncal antibody bevacizumab, which was evaluated for ovarian cancer treatment in clinical trials only recently. In the OCEANS study, a phase III randomized study, platinum-sensitive recurrent ovarian and fallopian tube cancer subjects (n=484) were randomized to the combination of gemcitabine and carboplatin either with or without bevacizumab for 6 to 10 cycles. The study showed that the combination therapy including administration of bevacizumab until disease progression resulted in a statistically significant improvement in progression-free survival (hazard ratio: 0.484, p<0.001) [146]. Bevacizumab was also evaluated as frontline therapy for patients in the randomized Phase III ICON7 trial, where stage IIIc or IV patients (n=1528) were randomized to carboplatin and paclitaxel with or without bevacizumab given concurrently every 3 weeks for 5 or 6 cycles and continued for 12 additional cycles or until progression of disease. The ICON7 study has demonstrated that bevacizumab improved the progression-free survival in women with high-risk ovarian cancer (hazard ratio: 0.84, p=0.004). The benefits with respect to both progression-free and overall survival were greater among those at high risk for disease progression [147].

Therefore, VEGF blockade can provide a) important antiangiogenic therapeutic effects, b) attenuate the endothelial blood-tumor barrier (Figure 3) and c) improve DC maturation. Tumor vascular endothelium is a physicial barrier through which T-cells home to the tumor and can present a significant challenge to the success of immunothery. Tumor endothelial cells regulate leukocyte trafficking via adhesion molecules and chemokines [148]. The adhesive properties of tumor endothelium can be deregulated by signaling through the endothelin-B receptor, resulting in the inability of T-cells to adhere and home effectively to tumors [105]. It is possible that some of these effects are maintained by VEGF, since VEGF blockade can enhance T-cell adhesion to endothelium and T-cell homing to tumors [149, 150]. Endothelial cells can also express surface inhibitory or death ligands mediators including PDL-1 and PDL-2 [151, 152], Fas ligand (FasL; also known as CD95L) [153], TNF-related apoptosis-inducing ligand (TRAIL) [154], and CD31 [155], or release soluble factors such IL-10, TGF-β and PGE2, which can inhibit effector lymphocyte function and/or DC maturation and function. It is quite possible that some of these tumor endothelial immunomodulatory mechanisms may not be directly mediated by VEGF. For example, we previoulsy found that many of the specific markers of tumor endothelium in ovarian cancer are induced not by VEGF but rather by a combination of hypoxia and inflammatory mediators [156]. Importantly, low dose cyclophosphamide used in this study to target Treg cells also exerts an antiangiogenic effect through direct cytotoxicity to tumor endothelium, which could synergize with bevacizumab to abate these aspects of the tumor endothelial barrier.

Figure 3
figure 3

VEGF blockade and the endothelial blood-tumor barrier.

VEGF plays an important role in suppression of DC maturation. It has been demonstrated that DCs from cancer patients are functionally impaired; furthermore, the increase of immature dendritic cells in the periphery was closely correlated with serum VEGF levels but not with TGF-β, IL-6 or GM-CSF [98]. Gabrilovich and colleagues were the first to identify that VEGF released by tumor cells were capable of impairing both DC function and DC maturation from CD34+ precursors [91]. By using neutralizing blocking antibodies against VEGF (but not IL-10 or TGF-β) they were able to reverse the suppression. VEGF can exert its immune suppression on dendritic cell mostly through disruption of normal hematopoesis [90, 94, 95] (impairment of normal nuclear factor-kappa-B signaling during hematopoesis [92] through VEGFR-1 [56]).

Treg cells have been shown to be present in ovarian cancer [24, 157]. Curiel et al. demonstrated that CD4+ CD25+ FOXP3+ human Treg cells suppress tumor-specific T-cell immunity and contribute to growth of human tumors in vivo in a study of 104 individuals affected with ovarian carcinoma. They found that there was a significant correlation between tumor Treg cell content and survival in the group as a, and also for individuals in stage II, , III or IV disease [158]. Presently, there are no specific drugs to deplete Treg cells, but among commercially available strategies, low-dose cyclophosphamide appears as a promising approach. In a preclinical rat model of colon cancer, Ghiringhelli et al. have shown that single administration of cyclophosphamide depletes CD4+ CD25+ T-cells, delays tumor growth, and improves cure rates when followed by non-curative immunotherapy [145]. The same group has demonstrated that metronomic oral cyclophosphamide immunosuppressive regulatory T-cells and improves effector immune function in patients with cancer [159]. Jaffee et al. showed that intravenous cyclophosphamide in doses no greater than 200 mg/m2 can transiently decrease Treg frequencies and enhance tumor-specific immune response in breast cancer patients in combination with a cancer vaccine [125, 160].

We recently reported on a pilot study (UPCC-11807), where we administered oral metronomic cyclophosphamide at 50 mg daily every other week to patients with EOC in combination with bevacizumab. We saw no significant effect of this dose of cyclophosphamide on peripheral blood Treg. Thus, in the present study we chose the dose of 200 mg/m2 administered intravenously, which conveniently matches the schedule of bevacizumab and vaccine. Based on reported effects, this schedule and dose of cyclophosphamide should reduce Treg cells. The concomitant administration of intravenous low-dose cyclophosphamide and bevacizumab has never been tested before. Importantly, Treg cells and VEGF are interconnected; hypoxia (which drives expression of VEGF) induces also accumulation of CCR10+ Treg cells in ovarian tumors, while Treg cells can in turn reprogram the tumor microenvironment towards angiogenesis [132]. Thus, in theory although VEGF blockade as monotherapy could attenuate the blood-tumor barrier, it could also produce a rebound increase in Treg accumulation in the tumor microenvironment, promoting tolerance and angiogenesis. In this case, concomitant suppression of Treg could deprive tumors from a critical homeostatic tolerance mechanism and could produce a synergistic immunomodulatory interaction at the tumor microenvironment, allowing a relatively weak antitumor immune response induced by cancer vaccine to become clinically effective. The growing understanding of these complex networks has revealed that the same cell populations or soluble factors can simultaneously promote angiogenesis and mediate immunosuppression in the tumor microenvironment, suggesting that successful cancer vaccine therapy may indeed benefit from effective blockade of multiple mechanisms [161]. We propose to block VEGF while also suppressing Treg using readily available FDA approved drugs, such as bevacizumab and low-dose cyclophosphamide (Figure 4).

Figure 4
figure 4

Combinatorial effect of VEGF blockade and Treg depletion.

In summary, this study responds to the urgent need created by the above observations to test in the clinic a combinatorial regimen that administers cancer vaccine in combination with Treg and VEGF blockade. The present trial will enable us to take the first step in this clinical development endeavor, testing the feasibility and safety of such an approach while we collect pilot biological data from the periphery and the tumor microenvironment. Upon completion of this phase I study, we will be in a position to dissect the individual contribution of bevacizumab or cyclophosphamide to vaccine therapy through rationally designed and adequately powered phase II randomized studies, based on the results of the present study.

Abbreviations

APC:

Antigen-presenting cell

CTL:

Cytotoxic T lymphocyte

CVPF:

Cell and vaccine production facility

DC:

Dendritic cell

DLT:

Dose-limiting toxicity

DTH:

Delayed type hypersensitivity

ECOG:

Eastern Cooperative Oncology Group

EOC:

Epithelial ovarian cancer

EOS:

End of study

GM-CSF:

Granulocyte/macrophage colony-stimulating factor

HOCL:

Hypochlorous acid

KLH:

Keyhole Limpet hemocyanin

LFTU:

Long-term follow-up

OCDC:

Oxidized tumor cell pulsed dendritic cell (vaccine)

PBMC:

Peripheral blood mononuclear cell

SAE:

Serious adverse event

TAA:

Tumor-associated antigen

TIL:

Tumor-infiltrating lymphocytes.

References

  1. Disis ML, Schiffman K: Cancer vaccines targeting the HER2/neu oncogenic protein. Semin Oncol. 2001, 28: 12-20.

    CAS  PubMed  Google Scholar 

  2. Hellstrom I, Goodman G, Pullman J, Yang Y, Hellstrom KE: Overexpression of HER-2 in ovarian carcinomas. Cancer Res. 2001, 61: 2420-2423.

    CAS  PubMed  Google Scholar 

  3. Murray JL, Przepiorka D, Ioannides CG: Clinical trials of HER-2/neu-specific vaccines. Semin Oncol. 2000, 27: 71-75. discussion 92–100

    CAS  PubMed  Google Scholar 

  4. Shih Ie M, Kurman RJ: Ovarian tumorigenesis: a proposed model based on morphological and molecular genetic analysis. Am J Pathol. 2004, 164: 1511-1518. 10.1016/S0002-9440(10)63708-X.

    PubMed  Google Scholar 

  5. Odunsi K, Jungbluth AA, Stockert E, Qian F, Gnjatic S, Tammela J, Intengan M, Beck A, Keitz B, Santiago D, Williamson B, Scanlan MJ, Ritter G, Chen YT, Driscoll D, Sood A, Lele S, Old LJ: NY-ESO-1 and LAGE-1 cancer-testis antigens are potential targets for immunotherapy in epithelial ovarian cancer. Cancer Res. 2003, 63: 6076-6083.

    CAS  PubMed  Google Scholar 

  6. Stockert E, Jager E, Chen YT, Scanlan MJ, Gout I, Karbach J, Arand M, Knuth A, Old LJ: A survey of the humoral immune response of cancer patients to a panel of human tumor antigens. J Exp Med. 1998, 187: 1349-1354. 10.1084/jem.187.8.1349.

    PubMed Central  CAS  PubMed  Google Scholar 

  7. Santomasso BD, Roberts WK, Thomas A, Williams T, Blachere NE, Dudley ME, Houghton AN, Posner JB, Darnell RB: A T cell receptor associated with naturally occurring human tumor immunity. Proc Natl Acad Sci USA. 2007, 104 (48): 19073-8. 10.1073/pnas.0704336104.

    PubMed Central  CAS  PubMed  Google Scholar 

  8. Counter CM, Hirte HW, Bacchetti S, Harley CB: Telomerase activity in human ovarian carcinoma. Proc Natl Acad Sci U S A. 1994, 91: 2900-2904. 10.1073/pnas.91.8.2900.

    PubMed Central  CAS  PubMed  Google Scholar 

  9. Vonderheide RH, Hahn WC, Schultze JL, Nadler LM: The telomerase catalytic subunit is a widely expressed tumor-associated antigen recognized by cytotoxic T lymphocytes. Immunity. 1999, 10: 673-679. 10.1016/S1074-7613(00)80066-7.

    CAS  PubMed  Google Scholar 

  10. Chen YT, Scanlan MJ, Sahin U, Tureci O, Gure AO, Tsang S, Williamson B, Stockert E, Pfreundschuh M, Old LJ: A testicular antigen aberrantly expressed in human cancers detected by autologous antibody screening. Proc Natl Acad Sci USA. 1997, 94: 1914-1918. 10.1073/pnas.94.5.1914.

    PubMed Central  CAS  PubMed  Google Scholar 

  11. Andersen MH: thor SP: Survivin–a universal tumor antigen. Histol Histopathol. 2002, 17: 669-675.

    CAS  PubMed  Google Scholar 

  12. Otto K, Andersen MH, Eggert A, Keikavoussi P, Pedersen LO, Rath JC, Bock M, Brocker EB, Straten PT, Kampgen E, Becker JC: Lack of toxicity of therapy-induced T cell responses against the universal tumour antigen survivin. Vaccine. 2005, 23: 884-889. 10.1016/j.vaccine.2004.08.007.

    CAS  PubMed  Google Scholar 

  13. Babcock B, Anderson BW, Papayannopoulos I, Castilleja A, Murray JL, Stifani S, Kudelka AP, Wharton JT, Ioannides CG: Ovarian and breast cytotoxic T lymphocytes can recognize peptides from the amino enhancer of split protein of the Notch complex. Mol Immunol. 1998, 35: 1121-1133. 10.1016/S0161-5890(98)00100-X.

    CAS  PubMed  Google Scholar 

  14. Peoples GE, Anderson BW, Fisk B, Kudelka AP, Wharton JT, Ioannides CG: Ovarian cancer-associated lymphocyte recognition of folate binding protein peptides. Ann Surg Oncol. 1998, 5: 743-750. 10.1007/BF02303486.

    CAS  PubMed  Google Scholar 

  15. Luby TM, Cole G, Baker L, Kornher JS, Ramstedt U, Hedley ML: Repeated immunization with plasmid DNA formulated in poly(lactide-co-glycolide) microparticles is well tolerated and stimulates durable T cell responses to the tumor-associated antigen cytochrome P450 1B1. Clin Immunol. 2004, 112: 45-53. 10.1016/j.clim.2004.04.002.

    CAS  PubMed  Google Scholar 

  16. Taylor-Papadimitriou J, Burchell J, Miles DW, Dalziel M: MUC1 and cancer. Biochim Biophys Acta. 1999, 1455: 301-313. 10.1016/S0925-4439(99)00055-1.

    CAS  PubMed  Google Scholar 

  17. Ghazizadeh M, Ogawa H, Sasaki Y, Araki T, Aihara K: Mucin carbohydrate antigens (T, Tn, and sialyl-Tn) in human ovarian carcinomas: relationship with histopathology and prognosis. Hum Pathol. 1997, 28: 960-966. 10.1016/S0046-8177(97)90012-5.

    CAS  PubMed  Google Scholar 

  18. Holmberg LA, Oparin DV, Gooley T, Lilleby K, Bensinger W, Reddish MA, MacLean GD, Longenecker BM, Sandmaier BM: Clinical outcome of breast and ovarian cancer patients treated with high-dose chemotherapy, autologous stem cell rescue and THERATOPE STn-KLH cancer vaccine. Bone Marrow Transplant. 2000, 25: 1233-1241. 10.1038/sj.bmt.1702430.

    CAS  PubMed  Google Scholar 

  19. Miles D, Papazisis K: Rationale for the clinical development of STn-KLH (Theratope) and anti-MUC-1 vaccines in breast cancer. Clin Breast Cancer. 2003, 3 (Suppl 4): S134-S138.

    PubMed  Google Scholar 

  20. Zhang L, Conejo-Garcia JR, Katsaros D, Gimotty PA, Massobrio M, Regnani G, Makrigiannakis A, Gray H, Schlienger K, Liebman MN, Rubin SC, Coukos G: Intratumoral T cells, recurrence, and survival in epithelial ovarian cancer. N Engl J Med. 2003, 348: 203-213. 10.1056/NEJMoa020177.

    CAS  PubMed  Google Scholar 

  21. Adams SF, Levine DA, Cadungog MG, Hammond R, Facciabene A, Olvera N, Rubin SC, Boyd J, Gimotty PA, Coukos G: Intraepithelial T cells and tumor proliferation: impact on the benefit from surgical cytoreduction in advanced serous ovarian cancer. Cancer. 2009, 115: 2891-2902. 10.1002/cncr.24317.

    PubMed Central  PubMed  Google Scholar 

  22. Clarke B, Tinker AV, Lee C, Subramanian S, Van De Rijn M, Turbin D, Kalloger S, Cadungog MG, Huntsman D, Coukos G, Gilks CB: Intraepithelial T cells and Prognosis in Ovarian Carcinoma: Novel Associations with Stage, Tumor Type and BRCA1 Loss. Mod Pathol. 2008, In Press

    Google Scholar 

  23. Hamanishi J, Mandai M, Iwasaki M, Okazaki T, Tanaka Y, Yamaguchi K, Higuchi T, Yagi H, Takakura K, Minato N, Honjo T, Fujii S: Programmed cell death 1 ligand 1 and tumor-infiltrating CD8+ T lymphocytes are prognostic factors of human ovarian cancer. Proc Natl Acad Sci USA. 2007, 104: 3360-3365. 10.1073/pnas.0611533104.

    PubMed Central  CAS  PubMed  Google Scholar 

  24. Sato E, Olson SH, Ahn J, Bundy B, Nishikawa H, Qian F, Jungbluth AA, Frosina D, Gnjatic S, Ambrosone C, Kepner J, Odunsi T, Ritter G, Lele S, Chen YT, Ohtani H, Old LJ, Odunsi K: Intraepithelial CD8+ tumor-infiltrating lymphocytes and a high CD8+/regulatory T cell ratio are associated with favorable prognosis in ovarian cancer. Proc Natl Acad Sci USA. 2005, 102: 18538-18543. 10.1073/pnas.0509182102.

    PubMed Central  CAS  PubMed  Google Scholar 

  25. Shah CA, Allison KH, Garcia RL, Gray HJ, Goff BA, Swisher EM, Shah CA, Allison KH, Garcia RL, Gray HJ, Goff BA, Swisher EM: Intratumoral T cells, tumor-associated macrophages, and regulatory T cells: Association with p53 mutations, circulating tumor DNA and survival in women with ovarian cancer. Gynecol Oncol. 2008, 109 (2): 215-9. 10.1016/j.ygyno.2008.01.010. Epub 2008 Mar 7

    CAS  PubMed  Google Scholar 

  26. Tomsova M, Melichar B, Sedlakova I, Steiner I: Prognostic significance of CD3+ tumor-infiltrating lymphocytes in ovarian carcinoma. Gynecol Oncol. 2008, 108: 415-420. 10.1016/j.ygyno.2007.10.016.

    CAS  PubMed  Google Scholar 

  27. Stumpf M, Hasenburg A, Riener MO, Jutting U, Wang C, Shen Y, Orlowska-Volk M, Fisch P, Wang Z, Gitsch G, Werner M, Lassmann S: Intraepithelial CD8-positive T lymphocytes predict survival for patients with serous stage III ovarian carcinomas: relevance of clonal selection of T lymphocytes. Br J Cancer. 2009, 101: 1513-1521. 10.1038/sj.bjc.6605274.

    PubMed Central  CAS  PubMed  Google Scholar 

  28. Milne K, Kobel M, Kalloger SE, Barnes RO, Gao D, Gilks CB, Watson PH, Nelson BH: Systematic analysis of immune infiltrates in high-grade serous ovarian cancer reveals CD20, FoxP3 and TIA-1 as positive prognostic factors. PLoS One. 2009, 4: e6412-10.1371/journal.pone.0006412.

    PubMed Central  PubMed  Google Scholar 

  29. Halapi E, Yamamoto Y, Juhlin C, Jeddi-Tehrani M, Grunewald J, Andersson R, Hising C, Masucci G, Mellstedt H, Kiessling R: Restricted T cell receptor V-beta and J-beta usage in T cells from interleukin-2-cultured lymphocytes of ovarian and renal carcinomas. Cancer Immunol Immunother. 1993, 36: 191-197. 10.1007/BF01741091.

    CAS  PubMed  Google Scholar 

  30. Hayashi K, Yonamine K, Masuko-Hongo K, Iida T, Yamamoto K, Nishioka K, Kato T: Clonal expansion of T cells that are specific for autologous ovarian tumor among tumor-infiltrating T cells in humans. Gynecol Oncol. 1999, 74: 86-92. 10.1006/gyno.1999.5430.

    CAS  PubMed  Google Scholar 

  31. Dadmarz RD, Ordoubadi A, Mixon A, Thompson CO, Barracchini KC, Hijazi YM, Steller MA, Rosenberg SA, Schwartzentruber DJ: Tumor-infiltrating lymphocytes from human ovarian cancer patients recognize autologous tumor in an MHC class II-restricted fashion. Cancer J Sci Am. 1996, 2: 263-272.

    CAS  PubMed  Google Scholar 

  32. Fisk B, Blevins TL, Wharton JT, Ioannides CG: Identification of an immunodominant peptide of HER-2/neu protooncogene recognized by ovarian tumor-specific cytotoxic T lymphocyte lines. J Exp Med. 1995, 181: 2109-2117. 10.1084/jem.181.6.2109.

    CAS  PubMed  Google Scholar 

  33. Kooi S, Freedman RS, Rodriguez-Villanueva J, Platsoucas CD: Cytokine production by T-cell lines derived from tumor-infiltrating lymphocytes from patients with ovarian carcinoma: tumor-specific immune responses and inhibition of antigen-independent cytokine production by ovarian tumor cells. Lymphokine Cytokine Res. 1993, 12: 429-437.

    CAS  PubMed  Google Scholar 

  34. Peoples GE, Goedegebuure PS, Smith R, Linehan DC, Yoshino I, Eberlein TJ: Breast and ovarian cancer-specific cytotoxic T lymphocytes recognize the same HER2/neu-derived peptide. Proc Natl Acad Sci USA. 1995, 92: 432-436. 10.1073/pnas.92.2.432.

    PubMed Central  CAS  PubMed  Google Scholar 

  35. Peoples GE, Schoof DD, Andrews JV, Goedegebuure PS, Eberlein TJ: T-cell recognition of ovarian cancer. Surgery. 1993, 114: 227-234.

    CAS  PubMed  Google Scholar 

  36. Santin AD, Bellone S, Ravaggi A, Pecorelli S, Cannon MJ, Parham GP: Induction of ovarian tumor-specific CD8+ cytotoxic T lymphocytes by acid-eluted peptide-pulsed autologous dendritic cells. Obstet Gynecol. 2000, 96: 422-430. 10.1016/S0029-7844(00)00916-9.

    CAS  PubMed  Google Scholar 

  37. Schlienger K, Chu CS, Woo EY, Rivers PM, Toll AJ, Hudson B, Maus MV, Riley JL, Choi Y, Coukos G, Kaiser LR, Rubin SC, Levine BL, Carroll RG, June CH: TRANCE- and CD40 ligand-matured dendritic cells reveal MHC class I-restricted T cells specific for autologous tumor in late-stage ovarian cancer patients. Clin Cancer Res. 2003, 9: 1517-1527.

    CAS  PubMed  Google Scholar 

  38. Edwards RP, Gooding W, Lembersky BC, Colonello K, Hammond R, Paradise C, Kowal CD, Kunschner AJ, Baldisseri M, Kirkwood JM, Herberman RB: Comparison of toxicity and survival following intraperitoneal recombinant interleukin-2 for persistent ovarian cancer after platinum: twenty-four-hour versus 7-day infusion. J Clin Oncol. 1997, 15: 3399-3407.

    CAS  PubMed  Google Scholar 

  39. Vlad AM, Budiu RA, Lenzner DE, Wang Y, Thaller JA, Colonello K, Crowley Nowick PA, Kelley JL, Price FV, Edwards RP: A phase II trial of intraperitoneal interleukin-2 in patients with platinum-resistant or platinum-refractory ovarian cancer. Cancer Immunol Immunother. 2010, 59 (2): 293-301. 10.1007/s00262-009-0750-3.

    CAS  PubMed  Google Scholar 

  40. Hodi FS, Butler M, Oble DA, Seiden MV, Haluska FG, Kruse A, Macrae S, Nelson M, Canning C, Lowy I, Korman A, Lautz D, Russell S, Jaklitsch MT, Ramaiya N, Chen TC, Neuberg D, Allison JP, Mihm MC, Dranoff G: Immunologic and clinical effects of antibody blockade of cytotoxic T lymphocyte-associated antigen 4 in previously vaccinated cancer patients. Proc Natl Acad Sci USA. 2008, 105: 3005-3010. 10.1073/pnas.0712237105.

    PubMed Central  CAS  PubMed  Google Scholar 

  41. Hodi FS, Mihm MC, Soiffer RJ, Haluska FG, Butler M, Seiden MV, Davis T, Henry-Spires R, MacRae S, Willman A, Padera R, Jaklitsch MT, Shankar S, Chen TC, Korman A, Allison JP, Dranoff G: Biologic activity of cytotoxic T lymphocyte-associated antigen 4 antibody blockade in previously vaccinated metastatic melanoma and ovarian carcinoma patients. Proc Natl Acad Sci USA. 2003, 100: 4712-4717. 10.1073/pnas.0830997100.

    PubMed Central  CAS  PubMed  Google Scholar 

  42. Odunsi K, Qian F, Matsuzaki J, Mhawech-Fauceglia P, Andrews C, Hoffman EW, Pan L, Ritter G, Villella J, Thomas B, Rodabaugh K, Lele S, Shrikant P, Old LJ, Gnjatic S: Vaccination with an NY-ESO-1 peptide of HLA class I/II specificities induces integrated humoral and T cell responses in ovarian cancer. Proc Natl Acad Sci USA. 2007, 104: 12837-12842. 10.1073/pnas.0703342104.

    PubMed Central  CAS  PubMed  Google Scholar 

  43. Mobus V, Horn S, Stock M, Schirrmacher V: Tumor cell vaccination for gynecological tumors. Hybridoma. 1993, 12: 543-547. 10.1089/hyb.1993.12.543.

    CAS  PubMed  Google Scholar 

  44. Hernando JJ, Park TW, Kubler K, Offergeld R, Schlebusch H, Bauknecht T: Vaccination with autologous tumour antigen-pulsed dendritic cells in advanced gynaecological malignancies: clinical and immunological evaluation of a phase I trial. Cancer Immunol Immunother. 2002, 51: 45-52. 10.1007/s00262-001-0255-1.

    CAS  PubMed  Google Scholar 

  45. Chu CS, Kim SH, June CH, Coukos G: Immunotherapy opportunities in ovarian cancer. Expert Rev Anticancer Ther. 2008, 8: 243-257. 10.1586/14737140.8.2.243.

    CAS  PubMed  Google Scholar 

  46. Leffers N, Daemen T, Helfrich W, Boezen HM, Cohlen BJ, Melief K, Nijman HW: Antigen-specific active immunotherapy for ovarian cancer. Cochrane Database Syst Rev. 2009, 4 (1): CD007287-10.1002/14651858.CD007287.pub2.

    Google Scholar 

  47. Chianese-Bullock KA, Irvin WP, Petroni GR, Murphy C, Smolkin M, Olson WC, Coleman E, Boerner SA, Nail CJ, Neese PY, Yuan A, Hogan KT, Slingluff CL: A multipeptide vaccine is safe and elicits T-cell responses in participants with advanced stage ovarian cancer. J Immunother. 2008, 31: 420-430. 10.1097/CJI.0b013e31816dad10.

    CAS  PubMed  Google Scholar 

  48. Tsuda N, Mochizuki K, Harada M, Sukehiro A, Kawano K, Yamada A, Ushijima K, Sugiyama T, Nishida T, Yamana H, Itoh K, Kamura T: Vaccination with predesignated or evidence-based peptides for patients with recurrent gynecologic cancers. J Immunother. 2004, 27 (1997): 60-72.

    CAS  PubMed  Google Scholar 

  49. Chu CS, Boyer J, Schullery DS, Gimotty PA, Gamerman V, Bender J, Levine BL, Coukos G, Rubin SC, Morgan MA, Vonderheide RH, June CH: Cancer Immunol Immunother. 2012, 61 (5): 629-41. 10.1007/s00262-011-1081-8. Epub 2011 Oct 22

    CAS  PubMed  Google Scholar 

  50. Chiang CL, Benencia F, Coukos G: Whole tumor antigen vaccines. Semin Immunol. 2010, 22: 132-143. 10.1016/j.smim.2010.02.004.

    PubMed Central  CAS  PubMed  Google Scholar 

  51. Toes RE, Schoenberger SP, van der Voort EI, Offringa R, Melief CJ: CD40-CD40Ligand interactions and their role in cytotoxic T lymphocyte priming and anti-tumor immunity. Semin Immunol. 1998, 10: 443-448. 10.1006/smim.1998.0147.

    CAS  PubMed  Google Scholar 

  52. Zajac AJ, Murali-Krishna K, Blattman JN, Ahmed R: Therapeutic vaccination against chronic viral infection: the importance of cooperation between CD4+ and CD8+ T cells. Curr Opin Immunol. 1998, 10: 444-449. 10.1016/S0952-7915(98)80119-2.

    CAS  PubMed  Google Scholar 

  53. Wong SB, Bos R, Sherman LA: Tumor-specific CD4+ T cells render the tumor environment permissive for infiltration by low-avidity CD8+ T cells. J Immunol. 2008, 180: 3122-3131.

    CAS  PubMed  Google Scholar 

  54. Cancer Genome Atlas Research N: Integrated genomic analyses of ovarian carcinoma. Nature. 2011, 474: 609-615. 10.1038/nature10166.

    Google Scholar 

  55. Neller MA, Lopez JA, Schmidt CW: Antigens for cancer immunotherapy. Semin Immunol. 2008, 20: 286-295. 10.1016/j.smim.2008.09.006.

    CAS  PubMed  Google Scholar 

  56. Dikov MM, Oyama T, Cheng P, Takahashi T, Takahashi K, Sepetavec T, Edwards B, Adachi Y, Nadaf S, Daniel T, Gabrilovich DI, Carbone DP: Vascular endothelial growth factor effects on nuclear factor-kappaB activation in hematopoietic progenitor cells. Cancer Res. 2001, 61: 2015-2021.

    CAS  PubMed  Google Scholar 

  57. Rabinowich H, Reichert TE, Kashii Y, Gastman BR, Bell MC, Whiteside TL: Lymphocyte apoptosis induced by Fas ligand- expressing ovarian carcinoma cells. Implications for altered expression of T cell receptor in tumor-associated lymphocytes. J Clin Invest. 1998, 101: 2579-2588. 10.1172/JCI1518.

    PubMed Central  CAS  PubMed  Google Scholar 

  58. Groh V, Wu J, Yee C, Spies T: Tumour-derived soluble MIC ligands impair expression of NKG2D and T-cell activation. Nature. 2002, 419: 734-738. 10.1038/nature01112.

    CAS  PubMed  Google Scholar 

  59. Prokopowicz ZM, Arce F, Biedron R, Chiang CL, Ciszek M, Katz DR, Nowakowska M, Zapotoczny S, Marcinkiewicz J, Chain BM: Hypochlorous acid: a natural adjuvant that facilitates antigen processing, cross-priming, and the induction of adaptive immunity. J Immunol. 2010, 184: 824-835. 10.4049/jimmunol.0902606.

    CAS  PubMed  Google Scholar 

  60. Deacon DH, Hogan KT, Swanson EM, Chianese-Bullock KA, Denlinger CE, Czarkowski AR, Schrecengost RS, Patterson JW, Teague MW, Slingluff CL: 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: 360-10.1186/1471-2407-8-360.

    PubMed Central  PubMed  Google Scholar 

  61. Coukos G, Courreges MC, Benencia F: Intraperitoneal oncolytic and tumor vaccination therapy with replication-competent recombinant virus: the herpes paradigm. Curr Gene Ther. 2003, 3: 113-125. 10.2174/1566523034578401.

    CAS  PubMed  Google Scholar 

  62. Chiang CL, Maier DA, Kandalaft LE, Brennan AL, Lanitis E, Ye Q, Levine BL, Czerniecki BJ, Powell DJ, Coukos G: Optimizing parameters for clinical-scale production of high IL-12 secreting dendritic cells pulsed with oxidized whole tumor cell lysate. J Transl Med. 2011, 9: 198-10.1186/1479-5876-9-198.

    PubMed Central  CAS  PubMed  Google Scholar 

  63. Callahan MK, Chaillot D, Jacquin C, Clark PR, Menoret A: Differential acquisition of antigenic peptides by Hsp70 and Hsc70 under oxidative conditions. J Biol Chem. 2002, 277: 33604-33609. 10.1074/jbc.M202890200.

    CAS  PubMed  Google Scholar 

  64. Stark JM: Immunological adjuvance of metabolic origin: oxidative stress, postulated impaired function of thiol proteases and immunogenicity. Scand J Immunol. 1998, 48: 475-479. 10.1046/j.1365-3083.1998.00443.x.

    CAS  PubMed  Google Scholar 

  65. Reth M: Hydrogen peroxide as second messenger in lymphocyte activation. Nat Immunol. 2002, 3: 1129-1134. 10.1038/ni1202-1129.

    CAS  PubMed  Google Scholar 

  66. Tatla S, Woodhead V, Foreman JC, Chain BM: The role of reactive oxygen species in triggering proliferation and IL-2 secretion in T cells. Free Radic Biol Med. 1999, 26: 14-24. 10.1016/S0891-5849(98)00133-6.

    CAS  PubMed  Google Scholar 

  67. Chiang CL, Ledermann JA, Aitkens E, Benjamin E, Katz DR, Chain BM: Oxidation of ovarian epithelial cancer cells by hypochlorous acid enhances immunogenicity and stimulates T cells that recognize autologous primary tumor. Clin Cancer Res. 2008, 14: 4898-4907. 10.1158/1078-0432.CCR-07-4899.

    CAS  PubMed  Google Scholar 

  68. Chiang CL, Ledermann JA, Rad AN, Katz DR, Chain BM: Hypochlorous acid enhances immunogenicity and uptake of allogeneic ovarian tumor cells by dendritic cells to cross-prime tumor-specific T cells. Cancer Immunol Immunother. 2006, 55: 1384-1395. 10.1007/s00262-006-0127-9.

    PubMed  Google Scholar 

  69. Anderson MM, Hazen SL, Hsu FF, Heinecke JW: Human neutrophils employ the myeloperoxidase-hydrogen peroxide-chloride system to convert hydroxy-amino acids into glycolaldehyde, 2-hydroxypropanal, and acrolein. A mechanism for the generation of highly reactive alpha-hydroxy and alpha,beta-unsaturated aldehydes by phagocytes at sites of inflammation. J Clin Invest. 1997, 99: 424-432. 10.1172/JCI119176.

    PubMed Central  CAS  PubMed  Google Scholar 

  70. Anderson MM, Requena JR, Crowley JR, Thorpe SR, Heinecke JW: The myeloperoxidase system of human phagocytes generates Nepsilon-(carboxymethyl)lysine on proteins: a mechanism for producing advanced glycation end products at sites of inflammation. J Clin Invest. 1999, 104: 103-113. 10.1172/JCI3042.

    PubMed Central  CAS  PubMed  Google Scholar 

  71. Hazen SL, Hsu FF, Mueller DM, Crowley JR, Heinecke JW: Human neutrophils employ chlorine gas as an oxidant during phagocytosis. J Clin Invest. 1996, 98: 1283-1289. 10.1172/JCI118914.

    PubMed Central  CAS  PubMed  Google Scholar 

  72. Carrasco-Marin E, Paz-Miguel JE, Lopez-Mato P, Alvarez-Dominguez C, Leyva-Cobian F: Oxidation of defined antigens allows protein unfolding and increases both proteolytic processing and exposes peptide epitopes which are recognized by specific T cells. Immunology. 1998, 95: 314-321. 10.1046/j.1365-2567.1998.00618.x.

    PubMed Central  CAS  PubMed  Google Scholar 

  73. Chen M, Masaki T, Sawamura T: LOX-1, the receptor for oxidized low-density lipoprotein identified from endothelial cells: implications in endothelial dysfunction and atherosclerosis. Pharmacol Ther. 2002, 95: 89-100. 10.1016/S0163-7258(02)00236-X.

    CAS  PubMed  Google Scholar 

  74. Moriwaki H, Kume N, Sawamura T, Aoyama T, Hoshikawa H, Ochi H, Nishi E, Masaki T, Kita T: Ligand specificity of LOX-1, a novel endothelial receptor for oxidized low density lipoprotein. Arterioscler Thromb Vasc Biol. 1998, 18: 1541-1547. 10.1161/01.ATV.18.10.1541.

    CAS  PubMed  Google Scholar 

  75. Oka K, Sawamura T, Kikuta K, Itokawa S, Kume N, Kita T, Masaki T: Lectin-like oxidized low-density lipoprotein receptor 1 mediates phagocytosis of aged/apoptotic cells in endothelial cells. Proc Natl Acad Sci USA. 1998, 95: 9535-9540. 10.1073/pnas.95.16.9535.

    PubMed Central  CAS  PubMed  Google Scholar 

  76. Xie J, Zhu H, Guo L, Ruan Y, Wang L, Sun L, Zhou L, Wu W, Yun X, Shen A, Gu J: Lectin-like oxidized low-density lipoprotein receptor-1 delivers heat shock protein 60-fused antigen into the MHC class I presentation pathway. J Immunol. 2010, 185: 2306-2313. 10.4049/jimmunol.0903214.

    CAS  PubMed  Google Scholar 

  77. Nestle FO, Alijagic S, Gilliet M, Sun Y, Grabbe S, Dummer R, Burg G, Schadendorf D: Vaccination of melanoma patients with peptide- or tumor lysate-pulsed dendritic cells. Nat Med. 1998, 4: 328-332. 10.1038/nm0398-328.

    CAS  PubMed  Google Scholar 

  78. Maier T, Tun-Kyi A, Tassis A, Jungius KP, Burg G, Dummer R, Nestle FO: Vaccination of patients with cutaneous T-cell lymphoma using intranodal injection of autologous tumor-lysate-pulsed dendritic cells. Blood. 2003, 102: 2338-2344. 10.1182/blood-2002-08-2455.

    CAS  PubMed  Google Scholar 

  79. Gitlitz BJ, Belldegrun AS, Zisman A, Chao DH, Pantuck AJ, Hinkel A, Mulders P, Moldawer N, Tso CL, Figlin RA: A pilot trial of tumor lysate-loaded dendritic cells for the treatment of metastatic renal cell carcinoma. J Immunother. 2003, 26: 412-419. 10.1097/00002371-200309000-00004.

    PubMed  Google Scholar 

  80. Iwashita Y, Tahara K, Goto S, Sasaki A, Kai S, Seike M, Chen CL, Kawano K, Kitano S: A phase I study of autologous dendritic cell-based immunotherapy for patients with unresectable primary liver cancer. Cancer Immunol Immunother. 2003, 52: 155-161.

    CAS  PubMed  Google Scholar 

  81. Thurner B, Roder C, Dieckmann D, Heuer M, Kruse M, Glaser A, Keikavoussi P, Kampgen E, Bender A, Schuler G: Generation of large numbers of fully mature and stable dendritic cells from leukapheresis products for clinical application. J Immunol Methods. 1999, 223: 1-15. 10.1016/S0022-1759(98)00208-7.

    CAS  PubMed  Google Scholar 

  82. Berger TG, Strasser E, Smith R, Carste C, Schuler-Thurner B, Kaempgen E, Schuler G: Efficient elutriation of monocytes within a closed system (Elutra) for clinical-scale generation of dendritic cells. J Immunol Methods. 2005, 298: 61-72. 10.1016/j.jim.2005.01.005.

    CAS  PubMed  Google Scholar 

  83. Berger TG, Feuerstein B, Strasser E, Hirsch U, Schreiner D, Schuler G, Schuler-Thurner B: Large-scale generation of mature monocyte-derived dendritic cells for clinical application in cell factories. J Immunol Methods. 2002, 268: 131-140. 10.1016/S0022-1759(02)00189-8.

    CAS  PubMed  Google Scholar 

  84. Ueno H, Schmitt N, Klechevsky E, Pedroza-Gonzalez A, Matsui T, Zurawski G, Oh S, Fay J, Pascual V, Banchereau J, Palucka K: Harnessing human dendritic cell subsets for medicine. Immunol Rev. 2010, 234: 199-212. 10.1111/j.0105-2896.2009.00884.x.

    PubMed Central  CAS  PubMed  Google Scholar 

  85. Ardavin C, Amigorena S, Reis E, Sousa C: Dendritic cells: immunobiology and cancer immunotherapy. Immunity. 2004, 20: 17-23. 10.1016/S1074-7613(03)00352-2.

    CAS  PubMed  Google Scholar 

  86. Banchereau J, Briere F, Caux C, Davoust J, Lebecque S, Liu YJ, Pulendran B, Palucka K: Immunobiology of dendritic cells. Annu Rev Immunol. 2000, 18: 767-811. 10.1146/annurev.immunol.18.1.767.

    CAS  PubMed  Google Scholar 

  87. Chiang CL, Hagemann AR, Leskowitz R, Mick R, Garrabrant T, Czerniecki BJ, Kandalaft LE, Powell DJ, Coukos G: Day-4 myeloid dendritic cells pulsed with whole tumor lysate are highly immunogenic and elicit potent anti-tumor responses. PLoS One. 2011, 6: e28732-10.1371/journal.pone.0028732.

    PubMed Central  CAS  PubMed  Google Scholar 

  88. Yang L, Carbone DP: Tumor-host immune interactions and dendritic cell dysfunction. Adv Cancer Res. 2004, 92: 13-27.

    CAS  PubMed  Google Scholar 

  89. Ohm JE, Carbone DP: VEGF as a mediator of tumor-associated immunodeficiency. Immunol Res. 2001, 23: 263-272. 10.1385/IR:23:2-3:263.

    CAS  PubMed  Google Scholar 

  90. Gabrilovich D, Ishida T, Oyama T, Ran S, Kravtsov V, Nadaf S, Carbone DP: Vascular endothelial growth factor inhibits the development of dendritic cells and dramatically affects the differentiation of multiple hematopoietic lineages in vivo. Blood. 1998, 92: 4150-4166.

    CAS  PubMed  Google Scholar 

  91. Gabrilovich DI, Chen HL, Girgis KR, Cunningham HT, Meny GM, Nadaf S, Kavanaugh D, Carbone DP: Production of vascular endothelial growth factor by human tumors inhibits the functional maturation of dendritic cells. Nat Med. 1996, 2: 1096-1103. 10.1038/nm1096-1096.

    CAS  PubMed  Google Scholar 

  92. Oyama T, Ran S, Ishida T, Nadaf S, Kerr L, Carbone DP, Gabrilovich DI: Vascular endothelial growth factor affects dendritic cell maturation through the inhibition of nuclear factor-kappa B activation in hemopoietic progenitor cells. J Immunol. 1998, 160: 1224-1232.

    CAS  PubMed  Google Scholar 

  93. Ishida T, Oyama T, Carbone DP, Gabrilovich DI: Defective function of Langerhans cells in tumor-bearing animals is the result of defective maturation from hemopoietic progenitors. J Immunol. 1998, 161: 4842-4851.

    CAS  PubMed  Google Scholar 

  94. Takahashi A, Kono K, Ichihara F, Sugai H, Fujii H, Matsumoto Y: Vascular endothelial growth factor inhibits maturation of dendritic cells induced by lipopolysaccharide, but not by proinflammatory cytokines. Cancer Immunol Immunother. 2004, 53: 543-550. 10.1007/s00262-003-0466-8.

    CAS  PubMed  Google Scholar 

  95. Ohm JE, Shurin MR, Esche C, Lotze MT, Carbone DP, Gabrilovich DI: Effect of vascular endothelial growth factor and FLT3 ligand on dendritic cell generation in vivo. J Immunol. 1999, 163: 3260-3268.

    CAS  PubMed  Google Scholar 

  96. Dikov MM, Ohm JE, Ray N, Tchekneva EE, Burlison J, Moghanaki D, Nadaf S, Carbone DP: Differential roles of vascular endothelial growth factor receptors 1 and 2 in dendritic cell differentiation. J Immunol. 2005, 174: 215-222.

    CAS  PubMed  Google Scholar 

  97. Curiel TJ, Wei S, Dong H, Alvarez X, Cheng P, Mottram P, Krzysiek R, Knutson KL, Daniel B, Zimmermann MC, David O, Burow M, Gordon A, Dhurandhar N, Myers L, Berggren R, Hemminki A, Alvarez RD, Emilie D, Curiel DT, Chen L, Zou W: Blockade of B7-H1 improves myeloid dendritic cell-mediated antitumor immunity. Nat Med. 2003, 9: 562-567. 10.1038/nm863.

    CAS  PubMed  Google Scholar 

  98. Almand B, Resser JR, Lindman B, Nadaf S, Clark JI, Kwon ED, Carbone DP, Gabrilovich DI: Clinical significance of defective dendritic cell differentiation in cancer. Clin Cancer Res. 2000, 6: 1755-1766.

    CAS  PubMed  Google Scholar 

  99. Gabrilovich DI, Ishida T, Nadaf S, Ohm JE, Carbone DP: Antibodies to vascular endothelial growth factor enhance the efficacy of cancer immunotherapy by improving endogenous dendritic cell function. Clin Cancer Res. 1999, 5: 2963-2970.

    CAS  PubMed  Google Scholar 

  100. Miyazaki A, Nakanishi T, Shimizu A, Mizobuchi M, Yamada Y, Imai K: Hb KOCHI [beta141(H19)Leu-->Val (g.1404-->C G); 144-->146(HC1-3)Lys-Tyr-His-->0 (g.1413 A-->T)]: a new variant with increased oxygen affinity. Hemoglobin. 2005, 29: 1-10.

    CAS  PubMed  Google Scholar 

  101. Nair S, Boczkowski D, Moeller B, Dewhirst M, Vieweg J, Gilboa E: Synergy between tumor immunotherapy and antiangiogenic therapy. Blood. 2003, 102: 964-971. 10.1182/blood-2002-12-3738.

    CAS  PubMed  Google Scholar 

  102. Li B, Lalani AS, Harding TC, Luan B, Koprivnikar K, Huan Tu G, Prell R, VanRoey MJ, Simmons AD, Jooss K: Vascular endothelial growth factor blockade reduces intratumoral regulatory T cells and enhances the efficacy of a GM-CSF-secreting cancer immunotherapy. Clin Cancer Res. 2006, 12: 6808-6816. 10.1158/1078-0432.CCR-06-1558.

    CAS  PubMed  Google Scholar 

  103. Li MO, Wan YY, Sanjabi S, Robertson AK, Flavell RA: Transforming growth factor-beta regulation of immune responses. Annu Rev Immunol. 2006, 24: 99-146. 10.1146/annurev.immunol.24.021605.090737.

    CAS  PubMed  Google Scholar 

  104. Rini BI, Weinberg V, Fong L, Conry S, Hershberg RM, Small EJ: Combination immunotherapy with prostatic acid phosphatase pulsed antigen-presenting cells (provenge) plus bevacizumab in patients with serologic progression of prostate cancer after definitive local therapy. Cancer. 2006, 107: 67-74. 10.1002/cncr.21956.

    CAS  PubMed  Google Scholar 

  105. Buckanovich RJ, Facciabene A, Kim S, Benencia F, Sasaroli D, Balint K, Katsaros D, O’Brien-Jenkins A, Gimotty PA, Coukos G: Endothelin B receptor mediates the endothelial barrier to T cell homing to tumors and disables immune therapy. Nat Med. 2008, 14: 28-36. 10.1038/nm1699.

    CAS  PubMed  Google Scholar 

  106. Kandalaft LE, Facciabene A, Buckanovich RJ, Coukos G: Endothelin B receptor, a new target in cancer immune therapy. Clin Cancer Res. 2009, 15: 4521-4528. 10.1158/1078-0432.CCR-08-0543.

    PubMed Central  CAS  PubMed  Google Scholar 

  107. Rosano L, Spinella F, Salani D, Di Castro V, Venuti A, Nicotra MR, Natali PG, Bagnato A: Therapeutic targeting of the endothelin a receptor in human ovarian carcinoma. Cancer Res. 2003, 63: 2447-2453.

    CAS  PubMed  Google Scholar 

  108. Spinella F, Rosano L, Elia G, Di Castro V, Natali PG, Bagnato A: Endothelin-1 Stimulates Cyclooxygenase-2 Expression in Ovarian Cancer Cells Through Multiple Signaling Pathways: Evidence for Involvement of Transactivation of the Epidermal Growth Factor Receptor. J Cardiovasc Pharmacol. 2004, 44: S140-S143. 10.1097/01.fjc.0000166255.12229.0d.

    CAS  PubMed  Google Scholar 

  109. Spinella F, Rosano L, Di Castro V, Decandia S, Nicotra MR, Natali PG, Bagnato A: Endothelin-1 and endothelin-3 promote invasive behavior via hypoxia-inducible factor-1alpha in human melanoma cells. Cancer Res. 2007, 67: 1725-1734. 10.1158/0008-5472.CAN-06-2606.

    CAS  PubMed  Google Scholar 

  110. Rosano L, Spinella F, Di Castro V, Nicotra MR, Albini A, Natali PG, Bagnato A: Endothelin receptor blockade inhibits molecular effectors of Kaposi’s sarcoma cell invasion and tumor growth in vivo. Am J Pathol. 2003, 163: 753-762. 10.1016/S0002-9440(10)63702-9.

    PubMed Central  CAS  PubMed  Google Scholar 

  111. Spinella F, Rosano L, Di Castro V, Natali PG, Bagnato A: Endothelin-1 induces vascular endothelial growth factor by increasing hypoxia-inducible factor-1alpha in ovarian carcinoma cells. J Biol Chem. 2002, 277: 27850-27855. 10.1074/jbc.M202421200.

    CAS  PubMed  Google Scholar 

  112. Salani D, Taraboletti G, Rosano L, Di Castro V, Borsotti P, Giavazzi R, Bagnato A: Endothelin-1 induces an angiogenic phenotype in cultured endothelial cells and stimulates neovascularization in vivo. Am J Pathol. 2000, 157: 1703-1711. 10.1016/S0002-9440(10)64807-9.

    PubMed Central  CAS  PubMed  Google Scholar 

  113. Zhang L, Yang N, Conejo-Garcia J-R, Mohamed A, Benencia F, Rubin SC, Allman D, Coukos G: Generation of a syngeneic mouse model to study the effects of vascular endothelial growth factor in ovarian carcinoma. Am J Pathol. 2002, 161: 2295-2309. 10.1016/S0002-9440(10)64505-1.

    PubMed Central  CAS  PubMed  Google Scholar 

  114. Manning EA, Ullman JG, Leatherman JM, Asquith JM, Hansen TR, Armstrong TD, Hicklin DJ, Jaffee EM, Emens LA: A vascular endothelial growth factor receptor-2 inhibitor enhances antitumor immunity through an immune-based mechanism. Clin Cancer Res. 2007, 13: 3951-3959. 10.1158/1078-0432.CCR-07-0374.

    CAS  PubMed  Google Scholar 

  115. Mendel DB, Laird AD, Smolich BD, Blake RA, Liang C, Hannah AL, Shaheen RM, Ellis LM, Weitman S, Shawver LK, Cherrington JM: Development of SU5416, a selective small molecule inhibitor of VEGF receptor tyrosine kinase activity, as an anti-angiogenesis agent. Anticancer Drug Des. 2000, 15: 29-41.

    CAS  PubMed  Google Scholar 

  116. Shrimali RK, Yu Z, Theoret MR, Chinnasamy D, Restifo NP, Rosenberg SA: Antiangiogenic agents can increase lymphocyte infiltration into tumor and enhance the effectiveness of adoptive immunotherapy of cancer. Cancer Res. 2010, 70: 6171-6180. 10.1158/0008-5472.CAN-10-0153.

    PubMed Central  CAS  PubMed  Google Scholar 

  117. Berd D, Mastrangelo MJ: Active immunotherapy of human melanoma exploiting the immunopotentiating effects of cyclophosphamide. Cancer Invest. 1988, 6: 337-349. 10.3109/07357908809080657.

    CAS  PubMed  Google Scholar 

  118. North RJ: Cyclophosphamide-facilitated adoptive immunotherapy of an established tumor depends on elimination of tumor-induced suppressor T cells. J Exp Med. 1982, 155: 1063-1074. 10.1084/jem.155.4.1063.

    CAS  PubMed  Google Scholar 

  119. Awwad M, North RJ: Cyclophosphamide-induced immunologically mediated regression of a cyclophosphamide-resistant murine tumor: a consequence of eliminating precursor L3T4+ suppressor T-cells. Cancer Res. 1989, 49: 1649-1654.

    CAS  PubMed  Google Scholar 

  120. Machiels JP, Reilly RT, Emens LA, Ercolini AM, Lei RY, Weintraub D, Okoye FI, Jaffee EM: Cyclophosphamide, doxorubicin, and paclitaxel enhance the antitumor immune response of granulocyte/macrophage-colony stimulating factor-secreting whole-cell vaccines in HER-2/neu tolerized mice. Cancer Res. 2001, 61: 3689-3697.

    CAS  PubMed  Google Scholar 

  121. Ercolini AM, Ladle BH, Manning EA, Pfannenstiel LW, Armstrong TD, Machiels JP, 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: 1591-1602. 10.1084/jem.20042167.

    PubMed Central  CAS  PubMed  Google Scholar 

  122. Leao IC, Ganesan P, Armstrong TD, Jaffee EM: Effective depletion of regulatory T cells allows the recruitment of mesothelin-specific CD8 T cells to the antitumor immune response against a mesothelin-expressing mouse pancreatic adenocarcinoma. Clin Transl Sci. 2008, 1: 228-239. 10.1111/j.1752-8062.2008.00070.x.

    PubMed Central  CAS  PubMed  Google Scholar 

  123. Curtis RE, Travis LB, Rowlings PA, Socie G, Kingma DW, Banks PM, Jaffe ES, Sale GE, Horowitz MM, Witherspoon RP, Shriner DA, Weisdorf DJ, Kolb HJ, Sullivan KM, Sobocinski KA, Gale RP, Hoover RN, Fraumeni JF, Deeg HJ: Risk of lymphoproliferative disorders after bone marrow transplantation: a multi-institutional study. Blood. 1999, 94: 2208-2216.

    CAS  PubMed  Google Scholar 

  124. Berd D, Maguire HC, Mastrangelo MJ: Induction of cell-mediated immunity to autologous melanoma cells and regression of metastases after treatment with a melanoma cell vaccine preceded by cyclophosphamide. Cancer Res. 1986, 46: 2572-2577.

    CAS  PubMed  Google Scholar 

  125. Emens LA, Asquith JM, Leatherman JM, Kobrin BJ, Petrik S, Laiko M, Levi J, Daphtary MM, Biedrzycki B, Wolff AC, Stearns V, Disis ML, Ye X, Piantadosi S, Fetting JH, Davidson NE, Jaffee EM: 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: 5911-5918. 10.1200/JCO.2009.23.3494.

    PubMed Central  CAS  PubMed  Google Scholar 

  126. Laheru D, Lutz E, Burke J, Biedrzycki B, Solt S, Onners B, Tartakovsky I, Nemunaitis J, Le D, Sugar E: 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: 1455-10.1158/1078-0432.CCR-07-0371.

    PubMed Central  CAS  PubMed  Google Scholar 

  127. Hahnfeldt P, Folkman J, Hlatky L: Minimizing long-term tumor burden: the logic for metronomic chemotherapeutic dosing and its antiangiogenic basis. J Theor Biol. 2003, 220: 545-554. 10.1006/jtbi.2003.3162.

    PubMed  Google Scholar 

  128. Garcia A, Oza A, Hirte H: Interim report of a phase II clinical trial of bevacizumab (Bev) and low dose metronomic oral cyclophophamide (mCTX) in recurrent ovarian (OC) and primary peritoneal carcinoma: a California Cancer Consortium Trial. J Clin Oncol. 2005, 23 (suppl): 5000-

    Google Scholar 

  129. Chura J, Van Iseghem K, Downs L, Carson LF, Judson P: Bevacizumab plus cyclophosphamide in heavily pretreated patients with recurrent ovarian cancer. Gynecol Oncol. 2007, 107: 326-330. 10.1016/j.ygyno.2007.07.017.

    CAS  PubMed  Google Scholar 

  130. Xu S, Koski GK, Faries M, Bedrosian I, Mick R, Maeurer M, Cheever MA, Cohen PA, Czerniecki BJ: Rapid high efficiency sensitization of CD8+ T cells to tumor antigens by dendritic cells leads to enhanced functional avidity and direct tumor recognition through an IL-12-dependent mechanism. J Immunol. 2003, 171: 2251-2261.

    CAS  PubMed  Google Scholar 

  131. Huang Y, Yuan J, Righi E, Kamoun WS, Ancukiewicz M, Nezivar J, Santosuosso M, Martin JD, Martin MR, Vianello F, Leblanc P, Munn LL, Huang P, Duda DG, Fukumura D, Jain RK, Poznansky MC: Vascular normalizing doses of antiangiogenic treatment reprogram the immunosuppressive tumor microenvironment and enhance immunotherapy. Proc Natl Acad Sci USA. 2012, 109: 17561-17566. 10.1073/pnas.1215397109.

    PubMed Central  CAS  PubMed  Google Scholar 

  132. Facciabene A, Peng X, Hagemann IS, Balint K, Barchetti A, Wang LP, Gimotty PA, Gilks CB, Lal P, Zhang L, Coukos G: Tumour hypoxia promotes tolerance and angiogenesis via CCL28 and T(reg) cells. Nature. 2011, 475: 226-230. 10.1038/nature10169.

    CAS  PubMed  Google Scholar 

  133. Marcinkiewicz J, Chain BM, Olszowska E, Olszowski S, Zgliczynski JM: Enhancement of immunogenic properties of ovalbumin as a result of its chlorination. Int J Biochem. 1991, 23: 1393-1395. 10.1016/0020-711X(91)90280-Z.

    CAS  PubMed  Google Scholar 

  134. Marcinkiewicz J, Olszowska E, Olszowski S, Zgliczynski JM: Enhancement of trinitrophenyl-specific humoral response to TNP proteins as the result of carrier chlorination. Immunology. 1992, 76: 385-388.

    PubMed Central  CAS  PubMed  Google Scholar 

  135. Engels FH, Koski GK, Bedrosian I, Xu S, Luger S, Nowell PC, Cohen PA, Czerniecki BJ: Calcium signaling induces acquisition of dendritic cell characteristics in chronic myelogenous leukemia myeloid progenitor cells. Proc Natl Acad Sci U S A. 1999, 96: 10332-10337. 10.1073/pnas.96.18.10332.

    PubMed Central  CAS  PubMed  Google Scholar 

  136. Czerniecki BJ, Cohen PA, Faries M, Xu S, Roros JG, Bedrosian I: Diverse functional activity of CD83+ monocyte-derived dendritic cells and the implications for cancer vaccines. Crit Rev Immunol. 2001, 21: 157-178.

    CAS  PubMed  Google Scholar 

  137. Koski GK, Cohen PA, Roses RE, Xu S, Czerniecki BJ: Reengineering dendritic cell-based anti-cancer vaccines. Immunol Rev. 2008, 222: 256-276. 10.1111/j.1600-065X.2008.00617.x.

    CAS  PubMed  Google Scholar 

  138. Butterfield LH, Disis ML, Fox BA, Lee PP, Khleif SN, Thurin M, Trinchieri G, Wang E, Wigginton J, Chaussabel D, Coukos G, Dhodapkar M, Hakansson L, Janetzki S, Kleen TO, Kirkwood JM, Maccalli C, Maecker H, Maio M, Malyguine A, Masucci G, Palucka AK, Potter DM, Ribas A, Rivoltini L, Schendel D, Seliger B, Selvan S, Slingluff CL, Stroncek DF, Streicher H, Wu X, Zeskind B, Zhao Y, Zocca MB, Zwierzina H, Marincola FM: A systematic approach to biomarker discovery; preamble to “the iSBTc-FDA taskforce on immunotherapy biomarkers”. J Transl Med. 2008, 6: 81-10.1186/1479-5876-6-81.

    PubMed Central  PubMed  Google Scholar 

  139. DeBenedette MA, Calderhead DM, Tcherepanova IY, Nicolette CA, Healey DG: Potency of mature CD40L RNA electroporated dendritic cells correlates with IL-12 secretion by tracking multifunctional CD8(+)/CD28(+) cytotoxic T-cell responses in vitro. J Immunother. 2011, 34: 45-57. 10.1097/CJI.0b013e3181fb651a.

    CAS  PubMed  Google Scholar 

  140. Watchmaker PB, Berk E, Muthuswamy R, Mailliard RB, Urban JA, Kirkwood JM, Kalinski P: Independent regulation of chemokine responsiveness and cytolytic function versus CD8+ T cell expansion by dendritic cells. J Immunol. 2010, 184: 591-597. 10.4049/jimmunol.0902062.

    PubMed Central  CAS  PubMed  Google Scholar 

  141. Okada H, Kalinski P, Ueda R, Hoji A, Kohanbash G, Donegan TE, Mintz AH, Engh JA, Bartlett DL, Brown CK, Zeh H, Holtzman MP, Reinhart TA, Whiteside TL, Butterfield LH, Hamilton RL, Potter DM, Pollack IF, Salazar AM, Lieberman FS: Induction of CD8+ T-cell responses against novel glioma-associated antigen peptides and clinical activity by vaccinations with {alpha}-type 1 polarized dendritic cells and polyinosinic-polycytidylic acid stabilized by lysine and carboxymethylcellulose in patients with recurrent malignant glioma. J Clin Oncol. 2011, 29: 330-336. 10.1200/JCO.2010.30.7744.

    PubMed Central  CAS  PubMed  Google Scholar 

  142. Lambert LA, Gibson GR, Maloney M, Durell B, Noelle RJ, Barth RJ: Intranodal immunization with tumor lysate-pulsed dendritic cells enhances protective antitumor immunity. Cancer Res. 2001, 61: 641-646.

    CAS  PubMed  Google Scholar 

  143. Bedrosian I, Mick R, Xu S, Nisenbaum H, Faries M, Zhang P, Cohen PA, Koski G, Czerniecki BJ: Intranodal administration of peptide-pulsed mature dendritic cell vaccines results in superior CD8+ T-cell function in melanoma patients. J Clin Oncol. 2003, 21: 3826-3835. 10.1200/JCO.2003.04.042.

    CAS  PubMed  Google Scholar 

  144. Yamamoto S, Konishi I, Mandai M, Kuroda H, Komatsu T, Nanbu K, Sakahara H, Mori T: Expression of vascular endothelial growth factor (VEGF) in epithelial ovarian neoplasms: correlation with clinicopathology and patient survival, and analysis of serum VEGF levels. Br J Cancer. 1997, 76: 1221-1227. 10.1038/bjc.1997.537.

    PubMed Central  CAS  PubMed  Google Scholar 

  145. Hartenbach EM, Olson TA, Goswitz JJ, Mohanraj D, Twiggs LB, Carson LF, Ramakrishnan S: Vascular endothelial growth factor (VEGF) expression and survival in human epithelial ovarian carcinomas. Cancer Lett. 1997, 121: 169-175. 10.1016/S0304-3835(97)00350-9.

    CAS  PubMed  Google Scholar 

  146. Aghajanian C, Blank SV, Goff BA, Judson PL, Teneriello MG, Husain A, Sovak MA, Yi J, Nycum LR: OCEANS: a randomized, double-blind, placebo-controlled phase III trial of chemotherapy with or without bevacizumab in patients with platinum-sensitive recurrent epithelial ovarian, primary peritoneal, or fallopian tube cancer. J Clin Oncol. 2012, 30: 2039-2045. 10.1200/JCO.2012.42.0505.

    PubMed Central  CAS  PubMed  Google Scholar 

  147. Perren TJ, Swart AM, Pfisterer J, Ledermann JA, Pujade-Lauraine E, Kristensen G, Carey MS, Beale P, Cervantes A, Kurzeder C, du Bois A, Sehouli J, Kimmig R, Stahle A, Collinson F, Essapen S, Gourley C, Lortholary A, Selle F, Mirza MR, Leminen A, Plante M, Stark D, Qian W, Parmar MK, Oza AM, Investigators I: A phase 3 trial of bevacizumab in ovarian cancer. N Engl J Med. 2011, 365: 2484-2496. 10.1056/NEJMoa1103799.

    CAS  PubMed  Google Scholar 

  148. Muller WA: Mechanisms of leukocyte transendothelial migration. Annu Rev Pathol. 2011, 6: 323-344. 10.1146/annurev-pathol-011110-130224.

    PubMed Central  CAS  PubMed  Google Scholar 

  149. Bouzin C, Brouet A, De Vriese J, Dewever J, Feron O: Effects of vascular endothelial growth factor on the lymphocyte-endothelium interactions: identification of caveolin-1 and nitric oxide as control points of endothelial cell anergy. J Immunol. 2007, 178: 1505-1511.

    CAS  PubMed  Google Scholar 

  150. Griffioen AW, Damen CA, Blijham GH, Groenewegen G: Tumor angiogenesis is accompanied by a decreased inflammatory response of tumor-associated endothelium. Blood. 1996, 88: 667-673.

    CAS  PubMed  Google Scholar 

  151. Mazanet MM, Hughes CC: B7-H1 is expressed by human endothelial cells and suppresses T cell cytokine synthesis. J Immunol. 2002, 169: 3581-3588.

    CAS  PubMed  Google Scholar 

  152. Rodig N, Ryan T, Allen JA, Pang H, Grabie N, Chernova T, Greenfield EA, Liang SC, Sharpe AH, Lichtman AH, Freeman GJ: Endothelial expression of PD-L1 and PD-L2 down-regulates CD8+ T cell activation and cytolysis. Eur J Immunol. 2003, 33: 3117-3126. 10.1002/eji.200324270.

    CAS  PubMed  Google Scholar 

  153. Sata M, Walsh K: TNFalpha regulation of Fas ligand expression on the vascular endothelium modulates leukocyte extravasation. Nat Med. 1998, 4: 415-420. 10.1038/nm0498-415.

    PubMed Central  CAS  PubMed  Google Scholar 

  154. Secchiero P, Zauli G: The puzzling role of TRAIL in endothelial cell biology. Arterioscler Thromb Vasc Biol. 2008, 28: e4-author reply e5-6

    CAS  PubMed  Google Scholar 

  155. Ma L, Mauro C, Cornish GH, Chai JG, Coe D, Fu H, Patton D, Okkenhaug K, Franzoso G, Dyson J, Nourshargh S, Marelli-Berg FM: Ig gene-like molecule CD31 plays a nonredundant role in the regulation of T-cell immunity and tolerance. Proc Natl Acad Sci USA. 2010, 107: 19461-19466. 10.1073/pnas.1011748107.

    PubMed Central  CAS  PubMed  Google Scholar 

  156. Sasaroli D, Gimotty PA, Pathak HB, Hammond R, Kougioumtzidou E, Katsaros D, Buckanovich R, Devarajan K, Sandaltzopoulos R, Godwin AK, Scholler N, Coukos G: Novel surface targets and serum biomarkers from the ovarian cancer vasculature. Cancer Biol Ther. 2011, 12: 169-180. 10.4161/cbt.12.3.16260.

    PubMed Central  CAS  PubMed  Google Scholar 

  157. Woo EY, Chu CS, Goletz TJ, Schlienger K, Yeh H, Coukos G, Rubin SC, Kaiser LR, June CH: Regulatory CD4(+)CD25(+) T cells in tumors from patients with early-stage non-small cell lung cancer and late-stage ovarian cancer. Cancer Res. 2001, 61: 4766-4772.

    CAS  PubMed  Google Scholar 

  158. Curiel TJ, Coukos G, Zou L, Alvarez X, Cheng P, Mottram P, Evdemon-Hogan M, Conejo-Garcia JR, Zhang L, Burow M, Zhu Y, Wei S, Kryczek I, Daniel B, Gordon A, Myers L, Lackner A, Disis ML, Knutson KL, Chen L, Zou W: Specific recruitment of regulatory T cells in ovarian carcinoma fosters immune privilege and predicts reduced survival. Nat Med. 2004, 10: 942-949. 10.1038/nm1093.

    CAS  PubMed  Google Scholar 

  159. Ghiringhelli F, Menard C, Puig PE, Ladoire S, Roux S, Martin F, Solary E, Le Cesne A, Zitvogel L, Chauffert B: Metronomic cyclophosphamide regimen selectively depletes CD4+CD25+ regulatory T cells and restores T and NK effector functions in end stage cancer patients. Cancer Immunol Immunother. 2007, 56: 641-648. 10.1007/s00262-006-0225-8.

    CAS  PubMed  Google Scholar 

  160. Le DT, Jaffee EM: Regulatory T-cell modulation using cyclophosphamide in vaccine approaches: a current perspective. Cancer Res. 2012, 72: 3439-3444. 10.1158/0008-5472.CAN-11-3912.

    PubMed Central  CAS  PubMed  Google Scholar 

  161. Motz GT, Coukos G: The parallel lives of angiogenesis and immunosuppression: cancer and other tales. Nat Rev Immunol. 2011, 11: 702-11. 10.1038/nri3064.

    CAS  PubMed  Google Scholar 

Download references

Acknowledgements

The development of the study protocol was supported by National Cancer Institute Ovarian SPORE grant P01-CA83638, National Institution of Health R01 grant FD003520-02, the Ovarian Cancer Immunotherapy Initiative, and the Marcus Foundation.

Author information

Authors and Affiliations

Authors

Corresponding author

Correspondence to Lana E Kandalaft.

Additional information

Competing interests

Authors declare that they do not have competing interest to disclose.

Authors’ contributions

LK contributed to the conception of the study, the clinical trial design, overseeing the clinical trial and writing the manuscript, CC contributed to the preclinical data. JT is the PI of the study, GM contributed to the preclinical data, KB contributed to writing the manuscript, RM contributed to the clinical trial design and the reviewing of the manuscript. GC contributed to the conception of the study, the clinical trial design, overseeing the clinical trial and reviewing the manuscript. All authors read and approved the final manuscript.

Authors’ original submitted files for images

Rights and permissions

This article is published under license to BioMed Central Ltd. This is an Open Access article distributed under the terms of the Creative Commons Attribution License (http://creativecommons.org/licenses/by/2.0), which permits unrestricted use, distribution, and reproduction in any medium, provided the original work is properly cited.

Reprints and permissions

About this article

Cite this article

Kandalaft, L.E., Chiang, C.L., Tanyi, J. et al. A Phase I vaccine trial using dendritic cells pulsed with autologous oxidized lysate for recurrent ovarian cancer. J Transl Med 11, 149 (2013). https://doi.org/10.1186/1479-5876-11-149

Download citation

  • Received:

  • Accepted:

  • Published:

  • DOI: https://doi.org/10.1186/1479-5876-11-149

Keywords