3,199
Views
46
CrossRef citations to date
0
Altmetric
Review

Immunotherapy in ovarian cancer

, &
Pages 1179-1191 | Received 01 Mar 2012, Accepted 13 May 2012, Published online: 21 Aug 2012

Abstract

Ovarian cancer is the most deadly gynecologic malignancy, with more than 15,000 deaths anticipated in 2012.1 While approximately 80% of patients will respond to frontline chemotherapy, more than 60% of patients will experience disease recurrence and only 44% will be alive at 5 years.1,2 Host anti-tumor immune responses are associated with a significant improvement in overall survival for women with ovarian cancer.3,4 By bolstering these responses, it may therefore be possible to significantly influence the prognosis of women with this lethal disease. In this review, we will focus on innovative immune-based strategies which are currently being investigated in the treatment of ovarian cancer.

Immunotherapy in Ovarian Cancer

Immunotherapeutic strategies in epithelial ovarian cancer have been a rising area of interest over the past two decades largely due to significant advancements in the knowledge of tumor antigens and antibody responses as well progression in the fields of cancer vaccines, lymphocyte transfer, and immunomodulatory therapy. It is now commonly believed that ovarian cancers are immunogenic tumors. A large stepping stone in the advancement of anti-tumor immune responses in ovarian carcinomas has been the characterization of tumor infiltrating lymphocytes (TILs).Citation5 Correlation between the presence of TILs and prolonged progression-free (PFS) and overall (OS) survival has been demonstrated in patients with advanced stage ovarian carcinoma,Citation4,Citation6 and the prognostic value of TILs was demonstrated to persist among all populations regardless of stage or grade of disease.Citation7 Specifically, the presence of CD8+ TILs has been demonstrated to correlate with increased survival.Citation6-Citation9 Confirmed by systematic review, CD8+ TILs are a superior marker for prognosis, as their presence correlates across all stages and histologies of ovarian carcinoma while CD3+ T cells only seem to show prognostic significance in serous ovarian carcinomas.Citation6 Adams, et al. reported that patients with more abundant CD8+ T cells demonstrated increased survival independent of tumor debulking, while patients with low CD8+ T cells showed significantly better prognosis if optimally debulked compared with those with suboptimal debulking.Citation3 These studies have resulted in an emerging consensus that, in the future, personalized therapy based on an individual’s immune prolife may alter outcome.

Conversely, the presence of immunosuppressive regulatory T cells (Tregs), classified as CD4+/CD25+/FoxP3+ T cells, have been associated with decreased survival in ovarian carcinoma.Citation10,Citation11 Woo, et al. were among the first to demonstrate increased proportions of CD4+CD25+ tumor associated Tregs, which secrete immunosuppressive TGF-β, in patients with advanced ovarian cancer.Citation12 Tregs have been found to inhibit nonspecific T cell activation in vitro and suppress endogenous tumor-associated antigen (TAA) specific T cell immunity. Curiel, et al. demonstrated an inverse correlation between the presence of Tregs and patient survival in ovarian cancers.Citation10 Sato, et al. further demonstrated that decreased survival occurs in patients with low ratios of CD8+/Tregs while high ratios of CD8+/Tregs are associated with increased survival. These data suggest that Tregs may have an adverse effect on the beneficial prognostic factors conferred by CD8+ TILs. Immune strategies targeting TILs are currently under investigation and will be discussed in detail below.

Additionally, ovarian cancers express tumor antigens, and patients have demonstrated spontaneous anti-tumor responses which are specific to these antigens.Citation8 A number of potential tumor antigens have been described in ovarian cancer with varying potential for vaccination strategies.Citation13 These antigens are separately classified as tumor-associated antigens (TAAs) and universal tumor antigens. TAAs can be sequestered from ascites or whole tumor collected during cytoreductive surgery. While TAAs can be specific to a patient and tumor, they are often also expressed by normal cells, creating limitations for their use. Currently several TAAs associated with ovarian cancer have been described and include HER2/neu, p53, CA125, STn, FR-α, mesothelin, NY-ESO-1, and cdr-2. Universal tumor antigens, including hTERT and survivin, are those expressed in a variety of tumors and are not found in most normal human cells. Immunotherapeutic regimens strengthening tumor antigen-specific anti-tumor responses have great potential in treating women with both recurrent and microscopic residual disease.

Despite promise for success, to date no advancement in the knowledge of tumor immunology has yielded a significant change in the standard therapy for ovarian carcinomas. The gold standard approach for these tumors is still a combination of cytoreductive surgery with carboplatin and paclitaxel. However, the immunogenicity of ovarian cancer yields great promise for future therapies.

Cancer Immunotherapy

Immunotherapy has found particular success in the treatment of other immunogenic cancers, in particular melanoma and renal cell carcinoma,Citation14 and successful strategies are being extrapolated into the treatment of ovarian cancer. Traditionally, immunotherapeutic strategies have focused on enhancing, inducing or suppressing innate or adaptive immune responses. Anti-tumor cytokines, including interferon-α (IFN-α), interferon-gamma (IFN-γ) and interleukin-1 (IL-1), as well as natural killer (NK) cells are targets for innate immune-based strategies. Adaptive-immune approaches aim to generate tumor antigen-specific cellular responses and include peptide vaccination, viral-based peptide vaccination, whole tumor antigen vaccination, anti-tumor monoclonal antibodies, and adoptive transfer of T lymphocytes and dendritic cells (DCs).Citation15 In addition, more recent approaches have investigated immunomodulatory strategies aimed at removing immune inhibitory responses due to Tregs and CTLA-4.Citation14,Citation15 ()

Table 1. Immunotherapeutic strategies under investigation in Ovarian Cancer

Cytokine therapy

Anti-tumor immune responses have been generated in preclinical models with the administration of cytokines, including IL-2, -4, -7, -12 and -18, IFN-α, IFN-γ, tumor necrosis factor α (TNF-α) and granulocyte-stimulating factor (GM-CSF).Citation16 Cytokine therapy affords the opportunity for immune regulation via induction and amplification of favorable antitumor immune responses.17 While cytokines are easy to manufacture and administer, they lack specific immunomodulatory effects.17

IL-2, a T-cell growth factor, has demonstrated anti-tumor responses in chemotherapy-resistant cancers, in particular melanoma and renal cell carcinoma.Citation18 Intraperitoneal (IP) IL-2 therapy was administered to women with platinum-resistant or -refractory ovarian cancer in a phase I/II trial.Citation18 Twenty-five percent experienced a treatment response with a median survival time of 2.1 y.Citation19 IL-2 treatment, which was well tolerated, resulted in a significant association between changes in CD3 T cells and IFN-γ producing CD8 T cell counts at early treatment time points and survival, suggesting that IP IL-2 should be further explored in the treatment of platinum-resistant ovarian cancer.

Interferon-α (IFN-α) administered at high doses can interfere with tumor cell replication, and at lower doses may activate T cells and upregulate MHC-I expression on ovarian cancer cells.Citation20 In a phase II trial, IP IFN-α alternating with cisplatin was administered to 14 women with minimal residual disease (≤ 5 mm) as salvage therapy.Citation21 Half of the cohort experienced pathologic complete remissions and remained disease free over a median follow-up of 22 mo (range 11–30 mo). In a subsequent phase I/II trial, IP IFN-α and carboplatin demonstrated an objective response of 42.8% in 16 women who had previously received intravenous cisplatin-based chemotherapy for recurrent or refractory ovarian cancer.Citation22

Interferon-gamma (IFN-γ) also exhibits several anti-tumor immune mechanisms, including upregulation of MHC expression on antigen presenting cells (APCs) and activation of T cell mediated pathways.Citation23 Preclinical evaluation of a CD80 (B7–1)/IFN-γ modified vaccine demonstrated enhancement of tumor-specific cytotoxic activity, resulting in reduction of tumor growth.Citation24 However, this vaccine has not yet been attempted in clinical trials.

Granulocyte-macrophage colony-stimulating factor (GM-CSF), a cytokine which stimulates the proliferation and differentiation of granulocytes and monocytes, has reported anti-tumor activity in a subset of cancer patients.Citation25 In a phase II trial, GM-CSF was administered to 72 asymptomatic patients with recurrent Müllerian malignancies without an indication for immediate systemic chemotherapy.Citation25 GM-CSF, which was generally well-tolerated, resulted in a decline in serum CA-125 levels which correlated with leukocytosis, as well as in a clinical response (1 complete response and 20 stable disease) in a subset of women.Citation25 GM-CSF was also evaluated in combination with recombinant interferon gamma 1b (rIFN-γ 1b) in a phase II trial of women with recurrent, platinum-sensitive ovarian, fallopian tube and primary peritoneal cancer.Citation26 In this cohort of 59 patients, GM-CSF and rIFN-γ 1b in combination with carboplatin produced a response rate of 56%.Citation26

In summary, cytokine immunotherapy has demonstrated promising results in ovarian cancer patients. However, these results need to be interpreted with caution due to the heterogeneity of the small study patient samples as well as the lack of standardization in measures of immunologic and clinical responses. Therefore, additional investigation is warranted to determine the clinical efficacy of these therapies.

Ovarian cancer vaccines

Peptide vaccines

Peptide vaccines target TAAs, including Her2/neu, NY-ESO-1, p53, VEGF and WT-1, which are expressed by ovarian cancer cells (). Peptide vaccines are generally well tolerated, easily produced, cost-effective, and have been shown to generate sustained immune responses.17 However, the immunogenicity of peptide vaccinations may require the co-administration of immune-boosting adjuvants, such as GM-CSF and/or Montanide to encourage induction of in vivo immune responses.Citation30 There are two subtypes of peptide vaccinations under investigation in ovarian cancer. Short-peptide vaccines aim to induce either T helper or cytotoxic T cell (CTL) responses via binding of epitopes specific for MHC I or II, respectively.Citation38 Long-peptide vaccines, unlike short-peptide vaccines, are not MHC restrictive and may contain epitopes to induce either T helper or CTL responses. These vaccines, however, have generally been less rigorously investigated in ovarian cancer patients.

Table 2. Peptide-based Vaccines studied in Ovarian Cancer

Her2/neu is a tumor antigen expressed in both breast and ovarian cancer. A vaccine containing MHC-II restricted HER-2/neu peptides was administered to patients who had HER-2/neu-overexpressing cancers.Citation27 The majority of patients developed antigen-specific IgG antibody immunityCitation27 as well as antigen-specific T cell responses.Citation28 These responses appeared to correlate with improved clinical outcomes.Citation28

NY-ESO-1 is a cancer-testis antigen which is highly immunogenic and has high expression in ovarian cancer.Citation39 In a phase I trial, 18 women with HLA-DP4+ epithelial ovarian cancer with minimal disease burden were administered a NY-ESO-1 peptide vaccine utilizing the epitope EOS157–170 with Montanide ISA51 adjuvant; this epitope is recognized by both HLA-DP4-restricted CD4+ T cells as well as HLA-A2 and HLA-A24 restricted CD8+ T cells.Citation29 This vaccine was well-tolerated and was able to generate both antigen specific CD4+ and CD8+ T cell responses. In a phase I trial, women with ovarian cancer in high-risk first remission were administered a HLA-A*0201-restricted NY-ESO-1b peptide vaccination with Montanide ISA-51 every 3 weeks for 5 vaccinations.Citation30 This vaccine was generally well-tolerated and capable of inducing specific CD8+ T-cell immunity in patients with or without NY-ESO-1 tumor expression. One third of patients experienced a complete clinical remission; however, these individuals had primary tumors which did not express NY-ESO-1, suggesting that NY-ESO-1 expression may be a dynamic process especially in patients with advanced disease.

Vaccination with subcutaneous short p53 peptide vs. intravenous p53 peptide pulsed DCs was recently compared in a phase II trial of women with stage III, IV or recurrent ovarian cancer.Citation31 Montanide and GM-CSF adjuvants were administered along with the subcutaneously injected peptide, while IL-2 was administered in both study groups. Both vaccination approaches resulted in comparable p53-specific immune responses with similar tolerability and safety, suggesting that the less demanding peptide vaccination strategy may be as effective as a cell-based approach.

A synthetic long-peptide (SLP) vaccine targeting p53 was evaluated in a phase II trial involving 20 patients with recurrent ovarian cancer after primary standard treatment.Citation32 The vaccine was generally well-tolerated and was successful in inducing p53-specific, Th2-dominant T cell responses in patients who received all four vaccinations. However, clinical response did not correlate with vaccine-mediated immunity. Further, p53-SLP vaccination did not have long-term impact on responses to secondary chemotherapy or survival in this cohort of patients.Citation33 In a second phase II trial, the authors combined the p53-SLP vaccine with cyclophosphamide with the aim of targeting Tregs and thereby improving the immunogenicity of the vaccine.Citation40 Combination therapy with cyclophosphamide and the p53-SLP vaccine induced higher p53-specific responses compared with p53-SLP monotherapy and produced a clinical response in 20% of patients, suggesting that further investigation is warranted.

Preclinical data suggests that immunization with a VEGF peptide generates a VEGF-specific antibody response which interferes with VEGF-dependent angiogenesis in an ovarian cancer xenograft nude mouse model.Citation41 Further investigation in humans is necessary to determine the role of this vaccine in treating or preventing ovarian cancer. A preliminary report has also suggested a possible role for WT1 peptide for treatment of recurrent ovarian cancer.Citation34

Multi-peptide vaccines have also been examined in ovarian cancer patients. A vaccine combining multiple MHC I restricted peptides present on ovarian cancer cells, including adenomatous polyposis coli (APC), ubiquitin conjugating enzyme E2, BAP31, replication protein A, Abl-binding protein 3c, Cyclin I, topoisomerase IIα, integrin β 8 subunit precursor, cell division control protein 2 (CDC2), TACE/ADAM17, g-catenin, and EDDR1, administered along with Montanide ISA-51 and GM-CSF was successful in generating peptide-specific T cell responses in patients with breast and ovarian cancer without evidence of disease.Citation42 However, 50% of the ovarian cancer patients experienced recurrence over a median follow-up of 492 d. An additional phase I trial examined the safety and immunogenicity of a vaccination with five MHC-1 restricted peptides and one MHC-2 restricted peptide along with Montanide ISA-51 and GM-CSF in nine women with epithelial ovarian, fallopian or primary peritoneal cancer who were HLA-A1, HLA-A2 or HLA-A3 positive.Citation43 Following in vitro stimulation, CD8+ T-cell responses were reported to the peptides in this vaccine, including folate binding protein (FBP191–199) and Her-2/neu754–762. While vaccine-related toxicities were low-grade, the vaccine lacked potency, did not demonstrate ex vivo immune responses, and failed to produce a substantial clinical effect.

In addition to peptide-based strategies, vaccines targeting sialyl-Tn (STn), a disaccharide molecule associated with MUC1, have been evaluated in phase I and II trials which included ovarian cancer patients.Citation36,Citation37 These vaccines were capable of inducing STn-specific immune responses but did not demonstrate a clinical benefit. Another carbohydrate epitope associated with MUC1, Lewisy, was examined in a phase I trial which included 25 ovarian cancer patients with complete clinical response to chemotherapy following primary therapy for residual or recurrent disease.Citation44 While this vaccine was generally well tolerated and was capable of inducing anti-Lewisy antibodies, a clinical benefit has yet to be determined.

Viral-based peptide vaccines

Recombinant viruses expressing human antigens have also been employed as vaccines in ovarian cancer patients. Antigens can be easily produced at high concentrations in vivo and these proteins are not HLA-restricted.Citation38 Intradermally injected recombinant vaccinia and fowlpox viruses expressing NY-ESO-1 were used to vaccinate 19 women with NY-ESO-1 expressing ovarian cancers who had complete responses to primary therapy.Citation45 This protocol was capable of inducing NY-ESO-1 specific immune responses in half of the patients, translating into a mean disease-free interval of 19.9 mo. Details of this trial have yet to be published. Recombinant vaccinia (PANVAC-V) and fowlpox (PANVAC-F) viruses producing both CEA and MUC1 as well as three T cell co-stimulatory molecules (B7.1, ICAM-1 and LFA-3) were administered along with GM-CSF in a phase I/II trial of 25 patients with progressive CEA or MUC1 overexpressing metastatic cancers (including three ovarian cancers).Citation46 This vaccination protocol was well-tolerated and was capable of inducing antigen-specific T cell responses. One of the three ovarian cancer patients achieved a durable clinical response lasting 18 mo following vaccination. Further investigation is necessary to determine the role of virus-based peptide vaccines in the treatment of ovarian cancer.

Whole tumor antigen vaccines

Unlike the above immunotherapies, whole tumor antigen vaccines have the potential to provide an individualized broad range of tumor antigens which may enhance host antigen-specific anti-tumor immune responses.Citation8 Whole tumor antigen vaccines can be created using tumor cells, autologous tumor lysates or tumor-derived RNA.Citation8 Given that they are generally poorly immunogenic due to a lack of stimulatory immune signals, whole tumor cell vaccines must be administered with adjuvants, such as GM-CSF, Montanide ISA-51 or Toll-like receptor agonists.Citation47 A recent meta-analysis suggests that individuals may have better clinical responses following whole tumor antigen vaccination compared with synthetic peptide vaccination.Citation48 A phase I/II randomized trial is currently underway at our institution which aims to determine the feasibility, safety and immunogenicity of an autologous oxidized tumor cell lysate vaccine (OC-L) in combination with Ampligen, a Toll-like receptor 3 agonist (NCT01312389).

In summary, ovarian cancer vaccines have been examined in several phase I and II clinical trials. Peptide vaccines, the most studied subtype, have resulted in encouraging clinical responses in ovarian cancer patients and are generally well tolerated and safe. However, peptide vaccines have poor immunogenicity requiring administration of immune adjuvants and have generally failed to demonstrate a clear clinical benefit. In contrast to peptide vaccines, viral-based peptide vaccines result in proteins which are not HLA restricted. Yet, preliminary studies have failed to report clinical responses with this type of vaccine. While offering the possibility of individualized treatment, the role of whole tumor antigen vaccines in ovarian cancer has not yet been determined. Further, this strategy relies on the availability of fresh tumor samples for vaccine development, and given the possibility of a broad range of antigens, it will likely be difficult to standardize vaccines among all ovarian cancer patients. However with continued investigation, vaccines may offer hope to ovarian cancer patients in both the therapeutic and preventive settings.

Monoclonal antibody treatment

Monoclonal antibody treatment aims to target antigens present on ovarian cancer cells. Traditionally, monoclonal antibodies have been used to facilitate an anti-tumor immune response via the neutralization of the tumor antigen, opsonization and activation of complement-mediated cytotoxicity, T cell-mediated cytotoxicity or antibody-dependent cell-mediated cytotoxicity.Citation14,Citation38 However, monoclonal antibodies can also be used to target molecules which are critical to tumor growth or survival pathways without eliciting an immune response.Citation20 Monoclonal antibody treatments have the advantages of being antigen-specific and generally easily to produce; however, in general, they are weakly immunogenic.17 Monoclonal antibodies targeting CA-125, MUC1, EpCAM, Her2/Neu, membrane folate receptor and VEGF have been examined in women with ovarian cancer. ()

Table 3. Monoclonal Antibodies studied in Ovarian Cancer

Immune-mediated monoclonal antibody treatment

CA125 (MUC-16) is a logical target for monoclonal antibody treatment given that 80% of ovarian cancers express this mucin.Citation49 Oregovomab (B43.13, OvaRex) is a monoclonal antibody to CA125 which has been examined in ovarian cancer patients in both phase II and III trials.Citation49,Citation50 In a prospective, open-label, pilot phase II trial involving heavily pretreated patients with ovarian cancer, antibody and T cell responses specific to both oregovomab and CA125 were generated in the majority of patients.Citation49 In three patients, these immune responses corresponded to stabilization of disease with survival greater than 2 y. In a subsequent trial of 20 women with advanced recurrent ovarian cancer, oregovomab was administered alone and then with optionally concurrent second-line chemotherapy.Citation50 Treatment-induced antibodies were generated, including human anti-mouse antibodies (HAMAs) and anti-oregovomab antibodies (Ab2) in 79% of patients and anti-CA125 antibodies in 11% of patients. T cell responses to CA125, generated in 39% of patients, and to autologous tumor (63% of patients) were significantly associated with improved survival (p = 0.002). Given that therapy was well tolerated and induced antigen-specific responses, subsequent trials examined the role of oregovomab in both frontlineCitation51 and maintenance therapy.Citation52 Preliminary results suggest that incorporation of oregovomab into frontline platinum/taxane chemotherapy may be more effective.Citation51

Abagovomab (ACA125) is an anti-idiotypic antibody targeting CA125 which has also been examined in ovarian cancer patients.Citation53,Citation54 Anti-idiotypic antibodies target primary anti-CA125 antibodies and boost the host immune response by mimicking the antigen of interest, producing anti-anti-idiotypic antibodies which also recognize this antigen.Citation15 In a phase Ib/II trial, abagovomab generated anti-anti-idiotypic antibodies and anti-CA125 antibodies in 68% and 50%, respectively, of 119 patients with advanced ovarian cancer.Citation54 Antibody-dependent cell-mediated cytotoxicity of CA125 positive cancer cells was observed in 26.9% of patients. Individuals who generated anti-anti-idiotypic antibodies experienced a significant survival advantage when compared with those who did not (p < 0.0001). Two other phase I trials also demonstrated robust anti-anti-idiotypic antibody responses to abagovomab but were unable to demonstrate similar clinical responses due to differences in study design.Citation53,Citation55

Given its overexpression in 90% of ovarian cancers, MUC1 is another promising target for monoclonal antibody therapy.Citation56 In a phase I trial of 26 women with persistent or recurrent ovarian cancer following platinum therapy, HMFG1, a murine anti-MUC1 antibody, resulted in a small but statistically significant rise in anti-HMFG1 and anti-MUC1 antibody responses in 38% of those completing the vaccination regimen.Citation56 However, while generally well-tolerated, HMFG1 lacked clinical efficacy in this trial. A radiolabeled form of this antibody, yttrium-90-muHMFG1, was also examined in subsequent phase I/II and III trials.Citation57,Citation58 In the phase I/II trial, yttrium-90-muHMFG1 administered intraperitoneally resulted in improved median survival in the women who received this antibody following traditional surgery and platinum-based chemotherapy.Citation57 However, despite a significant decrease in intraperitoneal disease following treatment, the phase III study failed to demonstrate a survival advantage due to increased extraperitoneal recurrences.Citation58

EpCAM is an antigen overexpressed in ovarian cancer, especially in patients with metastatic and recurrent/chemotherapy-resistant disease, and is therefore another attractive target for antibody-mediated immunotherapy.Citation59 Catumaxomab is a trifunctional antibody, targeting EpCAM on epithelial tumor cells and CD3 on T cells and is capable of recruiting and activating immune effector cells in the tumor microenvironment.Citation60 Preliminary results from recent phase II/III trials suggest that paracentesis plus intraperitoneal administration of catumaxomab may be beneficial in treating recurrent ovarian cancer patients with malignant ascites,Citation60,Citation61 with small but statistically significant increases in puncture-free survival and time to next paracentesis compared with patients undergoing paracentesis alone.

Non-immune-mediated monoclonal antibody treatment

Monoclonal antibodies which block tumor growth or survival pathways have also been examined in ovarian cancer patients. Recent data suggest that survival in ovarian cancer patients is inversely proportional to human epidermal growth factor receptor 2 (HER2) expression.Citation62 A monoclonal antibody targeting HER2, trastuzamab, is an available adjuvant therapy in HER2-positive breast cancer patients which has been recently examined in ovarian cancer patients. However, preliminary results in women with HER2-positive, recurrent or persistent ovarian or primary peritoneal cancers suggest a limited clinical benefit with an overall response rate of only 7.3%.Citation63 Monoclonal antibodies targeting FRα, including MOv18 and MORAb-003, have the potential of interrupting ovarian cancer cell growth by interfering with DNA synthesis.Citation64 Phase I trials in ovarian cancer patients have suggested that these antibodies are well tolerated but have limited clinical benefit in ovarian cancer patients.Citation65,Citation66

Bevacizumab, a humanized monoclonal antibody targeting VEGF-A, blocks angiogenesis which is a key pathway in tumorigenesis. Bevacizumab has been examined in ovarian cancer patients as a part of frontline therapyCitation67,Citation68 as well as secondary therapy for recurrent disease.Citation69,Citation70 Bevacizumab monotherapy demonstrated clinical response rates of 15–21% in two separate phase II trials of women with persistent or recurrent epithelial ovarian cancer, including those with platinum-resistant disease.Citation69,Citation70 Median PFS and OS ranged from 4.4 to 4.7 mo and 10.7 to 17 mo, respectively, and treatment was generally well-tolerated. However, Cannistra et al., reported a higher than expected rate of gastrointestinal perforation (11.4%), resulting in early termination of subject enrollment.

Recently, two large phase III randomized trials of bevacizumab in the frontline therapy of advanced ovarian cancer have been reported. ICON7 compared carboplatin/paclitaxel alone vs. carboplatin/paclitaxel/bevacizumab plus bevacizumab maintenance, and at 42 mo follow up, noted a 2 mo improvement in PFS (24.1 vs. 22.4 mo, p = 0.04), with benefit greatest for those at high risk for disease progression.Citation67 Similarly, the Gynecologic Oncology Group conducted a three arm randomized, double-blinded, placebo-controlled trial of carboplatin/paclitaxel with and without concurrent bevacizumab, with and without bevacizumab maintenance. The investigators noted a 4 mo improvement in PFS in those women receiving bevacizumab concurrently with primary chemotherapy as well as for maintenance (14.1 vs. 10.3 mo for combination therapy plus maintenance compared with carboplatin/paclitaxel alone, p < 0.001).Citation68 Given the high cost of bevacizumab, and the relatively modest influence on PFS, the role of its utility as a primary treatment modality in patients with advanced ovarian cancer remains unclear.

In summary, monoclonal antibody treatment is capable of producing antigen-specific immune responses in ovarian cancer patients. While results of phase I and II trials are promising, few randomized clinical trials have been conducted using this strategy, and the results of initial RCTs using anti-CA125 antibody therapy, the most studied target, have failed to demonstrated any clinical benefit despite inducing antigen specific immune responses.Citation71 Monoclonal antibodies targeting tumor growth and survival pathways, especially bevacizumab, have offered promising results in patients with recurrent disease. However, additional studies are needed to determine the role of these antibodies in frontline therapy.

Adoptive transfer of immune cells

Immune cells, including T cells, NK cells, DCs and macrophages, can be removed from a patient, manipulated ex vivo and then infused back into the same patient in order to boost anti-tumor cellular immune responses.Citation72

T cells

Adoptive transfer of autologous TILs has been met with success in treating metastatic melanoma patients, with objective response rates ranging up to 50%.Citation73 In a pilot trial, adoptive transfer of ex vivo IL-2 expanded TILs following a single dose of intravenous cyclophosphamide resulted in high response rates in a sample of women with advanced or recurrent ovarian cancer. Of the 7 patients treated with adoptive transfer of TILs alone, 1 achieved a complete response and 4 achieved partial responses.Citation74 In comparison, adoptive transfer of TILs in combination with cisplatin-based chemotherapy resulted in objective responses in 9 out of 10 patients (7 complete responses, 2 partial responses).

Since this study established the feasibility of adoptive T cell transfer, attention has been placed on optimizing the anti-tumor efficacy of this therapy. Given that the availability of tumor-reactive TILs is limited, investigators are looking into generating tumor-specific T cells via the ex vivo CD3/CD28-costimulation of vaccine-primed peripheral blood T cells or by genetically modifying peripheral blood T cells to express high affinity cloned T-cell receptors (TCRs) or chimeric antigen receptors (CARs).Citation8 In a phase I trial which included two patients with ovarian cancer, anti-CD3/anti-CD28 monoclonal antibody-co-activated T cells (COACTs) were capable of inducing cytokine production.Citation75 While this treatment was felt to be safe, the clinical benefit of this therapy has yet to be clearly defined. Our group is currently investigating the feasibility and safety of adoptive transfer using ex vivo CD3/CD28 co-stimulated autologous peripheral blood T cells in combination with a whole tumor lysates-pulsed dendritic cell vaccine (DCVax®-L) in patients with recurrent ovarian or primary peritoneal cancer (NCT00603460).

In a phase I trial of women with metastatic ovarian cancer, autologous T cells were genetically modified to express anti-FRα CAR.Citation76 While this therapy was generally well-tolerated, no clinical responses were demonstrated, likely due to poor localization of T cells into tumor as well a quick decline in circulating T cells. However, this was the first trial to explore the use of genetically modified T cells and additional studies are needed to further define the role in treating ovarian cancer patients.

Natural killer cells

NK cells are cytotoxic lymphocytes which are a part of the innate immune system and may play a role in tumor surveillance.Citation77 In a phase II trial which included 14 women with ovarian cancer, ex vivo activated haplo-identical related NK cells were adoptively transferred following lymphodepleting chemotherapy.Citation77 This protocol resulted in significant toxicities with reported transient donor chimerism and increased Treg responses. At a median of 36 d following transfer, 4 patients experienced a partial response while 12 had reported stable disease. Further investigation is warranted in order to determine the clinical benefit of this strategy in ovarian cancer.

Dendritic cells

DCs are specialized APCs which can be manipulated in order to boost tumor-antigen specific CTL responses via the recognition MHC-peptide complexes by peptide-specific TCRs on effector T cells.Citation78 DCs can present tumor antigens following exposure (or pulsing) to whole tumor cell lysates, tumor peptides or tumor cells.Citation79 A tumor cell lysate-pulsed dendritic cell vaccination was administered to women with advanced gynecologic malignancies in a recent phase I trial. Of the patients with progressive or recurrent ovarian cancer included in this trial, 50% demonstrated disease stabilization with PFS ranging 8–45 mo and lymphoproliferative responses were reported in two patients.Citation80 A phase I trial currently underway at our institution (NCT01132014) is examining the feasibility and immunogenicity of a DC vaccine loaded with autologous tumor lysate administered intranodally, alone or in combination with intravenous bevacizumab; results from this trial are still pending.

Our institution recently reported results of a randomized phase I/II trial of autologous DCs pulsed with Her-2/neu, hTERT, and PADRE peptides administered with or without low dose cyclophosphamide for 11 patients with advanced ovarian cancer in remission.Citation81 Of 9 patients receiving the full course of vaccinations, 3 recurred at 6, 17, and 6 mo, and 6 remain disease free at 36 mo of follow up. With no grade 3/4 vaccine-related toxicities noted, the 3-y overall survival was 90% with patients receiving cyclophosphamide demonstrating a non-significant survival advantage. Modest T cell responses to Her2/neu and hTERT were measured by IFN- γ ELISPOT, despite unexpected uniform baseline immunosuppression as demonstrated by virtually undetectable response to the diphtheria carrier protein component of the Prevnar™ vaccine. Brossart et al. conducted a trial of a HER-2/neu or MUC1-peptide pulsed autologous DC vaccine administered to ten patients with advanced breast or ovarian cancer.Citation82 In this pilot study, peptide-specific CTL responses were demonstrated in 50% of the patients; however, these responses were not correlated with clinical responses.

Schlienger et al. demonstrated that immature dendritic cells combined with autologous tumor cells could be matured using TNF-α and tumor necrosis factor-related activation-induced cytokine (TRANCE), and then used to induce tumor specific T cells capable of secreting IFN-γ.Citation5 A baseline T-cell response to autologous tumor associated antigens was measured in peripheral blood as well as the local tumor environment, indicating that tumor-specific T cells can be generated using autologous tumor cells as a source for antigens. Additional dendritic cell vaccines trials (NCT00703105, NCT00683241, and NCT01132014) are currently ongoing which may further elucidate a role for this technique in ovarian cancer patients.

In summary, cellular immunotherapy involving T cells, Natural Killer cells and Dendritic cells may be capable in boosting host anti-tumor cellular immune responses. However, this strategy is not well studied in ovarian cancer patients at this time, and therefore, results of these preliminary studies should be interpreted with caution.

Immunomodulation

Immunosuppressive responses have been reported in ovarian cancer patients which may limit the clinical effectiveness of the above proposed immunotherapeutic strategies.Citation83 Current investigations have focused on removing these immunologic brakes in order to facilitate host anti-tumor immune responses. As mentioned previously, Tregs are a subset of CD4+ T cells with immunosuppressive effects on host anti-tumor responses and are a poor prognostic indicator in ovarian cancer patients.Citation10,Citation11 Cyclophosphamide, an alkylating chemotherapeutic agent, has been used in an effort to reduce Treg responses. North and Berd demonstrated that cyclophosphamide eliminated CD8+ tumor suppressor cells, enhancing tumor immunotherapy and improving immune function.Citation84,Citation85 In animal models, cyclophosphamide administration has been linked to reduction in Tregs and enhancement of anti-tumor response.Citation86 However, initial studies in ovarian cancer patients demonstrated that cyclophosphamide failed to induce a reduction of Tregs or a qualitative difference in Treg function.Citation40,Citation81 Additional techniques to reduce Tregs focus on targeting CD25, the IL-2 receptor α chain, and include treatment with an anti-CD25 monoclonal antibody, Denileukin difitox and daclizumab.Citation8 While there are some promising results from pilot studies with melanoma or breast patients, the impact of these CD25 targeting therapies have not been fully delineated in ovarian cancer patients.

T cell activation requires that TCRs on T cells recognize peptides presented by MHC-I or MHC-II complexes on APCs. This process is regulated by stimulatory or inhibitory receptors present on T cells. The latter, including CTLA-4 and programmed death 1 (PD-1), can significantly dampen effector T cell responses.Citation8 By blocking these receptors, it may therefore be feasible to enhance the clinical benefit of spontaneous anti-tumor immune responses or those brought on by immunotherapeutic treatment.

Ipilimumab is a monoclonal antibody targeting CTLA-4 which has demonstrated promising clinical benefit in patients with melanoma and renal cell carcinoma.Citation14 In a phase III trial, ipilimumab provided a significant survival advantage in patients with previously treated metastatic melanoma albeit with some pronounced toxicity.Citation87 The same investigators then expanded their results to include the treatment of ovarian cancer patients.Citation83,Citation88 This group treated a total of eleven patients with advanced ovarian cancer using modified autologous tumor cells which secrete GM-CSF (GVAX) followed by ipilimumab.Citation83,Citation88 In several patients in this cohort, ipilimumab demonstrated significant anti-tumor effects corresponding with a drop in serum CA125 levels. These preliminary results suggest that CTLA-4 may be an appropriate target in ovarian cancer patients. PD-1 has been targeted in phase I trials with patients with hematologic malignancies and has yet to be examined in women with ovarian cancer.Citation8 This inhibitory receptor seems like another justifiable target in ovarian cancer given that its receptor PD-L1 is expressed on ovarian cancer cells.Citation8

Traditional cytotoxic agents, most notably pegylated liposomal doxorubicin (PLD), may also have immunomodulatory effects on ovarian cancer cells.Citation89 In addition to causing direct DNA damage, PLD alters the immunophenotype of surviving ovarian tumor cells and renders them more susceptible to anti-tumor T cell mediated cytotoxicity. Preliminary results demonstrated in preclinical studies may justify the combination of PLD with immunotherapeutic strategies aiming to enhance anti-tumor T cell responses. However, these findings have not yet been confirmed in patients with ovarian cancer.

Immunomodulation may be prove to be the key to improving clinical responses in ovarian cancer patients undergoing immunotherapeutic treatment. While there are some promising results from pilot studies with melanoma or breast patients, the impact of these strategies have not been fully examined in ovarian cancer patients

Immunotherapy in Ovarian Cancer: Critical Commentary

Innovative immunotherapeutic strategies offer the promise of enhancing host anti-tumor responses which may improve clinical outcomes in women with ovarian cancer. While many preliminary phase I/II studies have demonstrated induction of anti-tumor responses, there is current no clinically effective antigen-specific active immunotherapy available for women with ovarian cancer.Citation71

One major pitfall highlighted by this review is the examination of immunotherapeutics in women with recurrent and widely metastatic disease. These individuals likely have heavy tumor burden as a byproduct of tumor immune-evading responses, and this may account for the general lack of clinical benefit seen thus far with investigated immunotherapeutic strategies in ovarian cancer.Citation38 Immunomodulatory techniques combined with previously examined immunotherapies offer the promise of improving clinical responses in those with recurrent disease, and we believe strategies targeting Tregs and inhibitory signals such as CTLA-4 are most likely to be fruitful. Further, immunotherapy has been rarely studied in women with primary disease. While we appreciate that immunotherapy is unlikely to replace standard adjuvant chemotherapy, we believe that further investigation is warranted to determine whether immunotherapy could be combined with frontline therapy or administered as maintenance or consolidation therapy.

Additionally, we believe that it is important to select appropriate candidates for clinical trials examining immunotherapy in ovarian cancer. Candidates for immunotherapy would ideally have documented baseline immune responses which would render them more responsive to these strategies. For example, ovarian cancer patients with naturally occurring tumor antigen-specific antibody responses and/or those whose tumors express tumor-associated antigens or contain abundant TILs may be more likely to demonstrate a clinical response to immunotherapies. Conversely, patients who have underlying autoimmunity may have aberrant immune responses interfering with the generation of appropriate anti-tumor responses, making them less appropriate candidates for immunotherapy. In the future, we expect that immunotherapeutic regimens will be offered to individuals with favorable immune biomarker profiles which may translate into improved prognostic outcomes.

Ongoing investigation may help to define the role of immunotherapy alone or in combination with synergistic treatment strategies in the treatment of ovarian cancer patients (). With careful selection of candidates as well as appropriate reporting of standardized treatment responses and adverse events, these trials may determine a role for immunotherapy in the treatment of ovarian cancer.

Table 4. Current Clinical Trials investigating Immunotherapy in Ovarian Cancer

Abbreviations:
TILs=

tumor infiltrating lymphocytes

Tregs=

T regulatory cells

TGF-β=

ransforming growth factor-beta

FR-α=

folate receptor-alpha

IFN-α=

Interferon-alpha

IFN-γ=

interferon-gamma

IL-1=

Interleukin-1

NK=

natural killer

DCs=

dendritic cells

MDSCs=

myeloid-derived suppressor cells

TNF-α=

tumor necrosis factor-alpha

GM-CSF=

granulocyte-macrophage colony-stimulating factor

IP=

intraperitoneal

MHC=

major histocompatibility complex

APCs=

antigen presenting cells

r IFN-γ 1b=

recombinant interferon gamma 1b

VEGF=

vascular endothelial growth factor

WT-1=

Wilms tumor-1

CTLs=

cytotoxic T lymphocytes

SLP=

synthetic long-peptide

FBP=

folate binding protein

STn=

sialyl-Tn

HAMAs=

human anti-mouse antibodies

EpCAM=

epithelial cell adhesion molecule

HER2=

human epidermal growth factor receptor 2

TCR=

T cell receptor

CARs=

chimeric antigen receptors

COACTs=

anti-CD3/anti-CD28 monoclonal antibody-coactivated T cells

TRANCE=

tumor necrosis factor-related activation-induced cytokine

PD-1=

programmed death 1

CTLA-4=

cytotoxic T lymphocyte-associated antigen 4

OS=

overall survival

PFS=

progression-free survival

Acknowledgments

We would like to thank our colleagues for their thoughtful review of this manuscript.

Disclosure of Potential Conflicts of Interest

The authors attest that there are no reportable conflicts of interest with the publication of the manuscript.

References

  • Siegel R, Naishadham D, Jemal A. Cancer statistics, 2012. CA Cancer J Clin 2012; 62:10 - 29; http://dx.doi.org/10.3322/caac.20138; PMID: 22237781
  • Cannistra SA. Cancer of the ovary. N Engl J Med 2004; 351:2519 - 29; http://dx.doi.org/10.1056/NEJMra041842; PMID: 15590954
  • Adams SF, Levine DA, Cadungog MG, Hammond R, Facciabene A, Olvera N, et al. Intraepithelial T cells and tumor proliferation: impact on the benefit from surgical cytoreduction in advanced serous ovarian cancer. Cancer 2009; 115:2891 - 902; http://dx.doi.org/10.1002/cncr.24317; PMID: 19472394
  • Zhang L, Conejo-Garcia JR, Katsaros D, Gimotty PA, Massobrio M, Regnani G, et al. Intratumoral T cells, recurrence, and survival in epithelial ovarian cancer. N Engl J Med 2003; 348:203 - 13; http://dx.doi.org/10.1056/NEJMoa020177; PMID: 12529460
  • Schlienger K, Chu CS, Woo EY, Rivers PM, Toll AJ, Hudson B, et al. 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 - 27; PMID: 12684428
  • Clarke B, Tinker AV, Lee CH, Subramanian S, van de Rijn M, Turbin D, et al. Intraepithelial T cells and prognosis in ovarian carcinoma: novel associations with stage, tumor type, and BRCA1 loss. Mod Pathol 2009; 22:393 - 402; http://dx.doi.org/10.1038/modpathol.2008.191; PMID: 19060844
  • Hwang WT, Adams SF, Tahirovic E, Hagemann IS, Coukos G. Prognostic significance of tumor-infiltrating T cells in ovarian cancer: a meta-analysis. Gynecol Oncol 2012; 124:192 - 8; http://dx.doi.org/10.1016/j.ygyno.2011.09.039; PMID: 22040834
  • Kandalaft LE, Powell DJ Jr., Singh N, Coukos G. Immunotherapy for ovarian cancer: what’s next?. J Clin Oncol 2011; 29:925 - 33; http://dx.doi.org/10.1200/JCO.2009.27.2369; PMID: 21079136
  • Leffers N, Fehrmann RS, Gooden MJ, Schulze UR, Ten Hoor KA, Hollema H, et al. Identification of genes and pathways associated with cytotoxic T lymphocyte infiltration of serous ovarian cancer. Br J Cancer 2010; 103:685 - 92; http://dx.doi.org/10.1038/sj.bjc.6605820; PMID: 20664601
  • Curiel TJ, Coukos G, Zou L, Alvarez X, Cheng P, Mottram P, et al. Specific recruitment of regulatory T cells in ovarian carcinoma fosters immune privilege and predicts reduced survival. Nat Med 2004; 10:942 - 9; http://dx.doi.org/10.1038/nm1093; PMID: 15322536
  • Sato E, Olson SH, Ahn J, Bundy B, Nishikawa H, Qian F, et al. 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 U S A 2005; 102:18538 - 43; http://dx.doi.org/10.1073/pnas.0509182102; PMID: 16344461
  • Woo EY, Chu CS, Goletz TJ, Schlienger K, Yeh H, Coukos G, et al. 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 - 72; PMID: 11406550
  • Chu CS, Kim SH, June CH, Coukos G. Immunotherapy opportunities in ovarian cancer. Expert Rev Anticancer Ther 2008; 8:243 - 57; http://dx.doi.org/10.1586/14737140.8.2.243; PMID: 18279065
  • Topalian SL, Weiner GJ, Pardoll DM. Cancer immunotherapy comes of age. J Clin Oncol 2011; 29:4828 - 36; http://dx.doi.org/10.1200/JCO.2011.38.0899; PMID: 22042955
  • Liu B, Nash J, Runowicz C, Swede H, Stevens R, Li Z. Ovarian cancer immunotherapy: opportunities, progresses and challenges. J Hematol Oncol 2010; 3:7; http://dx.doi.org/10.1186/1756-8722-3-7; PMID: 20146807
  • Connor J, Bannerji R, Saito S, Heston W, Fair W, Gilboa E. Regression of bladder tumors in mice treated with interleukin 2 gene-modified tumor cells. J Exp Med 1993; 177:1127 - 34; http://dx.doi.org/10.1084/jem.177.4.1127; PMID: 8459207
  • Liu B, Nash J, Runowicz C, Swede H, Stevens R, Li Z. Ovarian cancer immunotherapy: opportunities, progresses and challenges. J Hematol Oncol 2010; 3:7; http://dx.doi.org/10.1186/1756-8722-3-7; PMID: 20146807
  • Edwards RP, Gooding W, Lembersky BC, Colonello K, Hammond R, Paradise C, et al. 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 - 407; PMID: 9363872
  • Vlad AM, Budiu RA, Lenzner DE, Wang Y, Thaller JA, Colonello K, et al. A phase II trial of intraperitoneal interleukin-2 in patients with platinum-resistant or platinum-refractory ovarian cancer. Cancer Immunol Immunother 2010; 59:293 - 301; http://dx.doi.org/10.1007/s00262-009-0750-3; PMID: 19690855
  • Thibodeaux SR, Curiel TJ. Immune therapy for ovarian cancer: promise and pitfalls. Int Rev Immunol 2011; 30:102 - 19; http://dx.doi.org/10.3109/08830185.2011.567361; PMID: 21557637
  • Nardi M, Cognetti F, Pollera CF, Giulia MD, Lombardi A, Atlante G, et al. Intraperitoneal recombinant alpha-2-interferon alternating with cisplatin as salvage therapy for minimal residual-disease ovarian cancer: a phase II study. J Clin Oncol 1990; 8:1036 - 41; PMID: 2189953
  • Repetto L, Chiara S, Guido T, Bruzzone M, Oliva C, Ragni N, et al. Intraperitoneal chemotherapy with carboplatin and interferon alpha in the treatment of relapsed ovarian cancer: a pilot study. Anticancer Res 1991; 11:1641 - 3; PMID: 1746921
  • Kaur T, Slavcev RA, Wettig SD. Addressing the challenge: current and future directions in ovarian cancer therapy. Curr Gene Ther 2009; 9:434 - 58; http://dx.doi.org/10.2174/156652309790031148; PMID: 20021329
  • Qian HN, Liu GZ, Cao SJ, Feng J, Ye X. The experimental study of ovarian carcinoma vaccine modified by human B7-1 and IFN-gamma genes. Int J Gynecol Cancer 2002; 12:80 - 5; http://dx.doi.org/10.1046/j.1525-1438.2002.01060.x; PMID: 11860540
  • Roche MR, Rudd PJ, Krasner CN, Matulonis UA, Berlin ST, Lee H, et al. Phase II trial of GM-CSF in women with asymptomatic recurrent müllerian tumors. Gynecol Oncol 2010; 116:168 - 72; http://dx.doi.org/10.1016/j.ygyno.2009.10.075; PMID: 19922985
  • Schmeler KM, Vadhan-Raj S, Ramirez PT, Apte SM, Cohen L, Bassett RL, et al. A phase II study of GM-CSF and rIFN-gamma1b plus carboplatin for the treatment of recurrent, platinum-sensitive ovarian, fallopian tube and primary peritoneal cancer. Gynecol Oncol 2009; 113:210 - 5; http://dx.doi.org/10.1016/j.ygyno.2009.02.007; PMID: 19264351
  • Disis ML, Goodell V, Schiffman K, Knutson KL. Humoral epitope-spreading following immunization with a HER-2/neu peptide based vaccine in cancer patients. J Clin Immunol 2004; 24:571 - 8; http://dx.doi.org/10.1023/B:JOCI.0000040928.67495.52; PMID: 15359116
  • Disis ML, Gooley TA, Rinn K, Davis D, Piepkorn M, Cheever MA, et al. Generation of T-cell immunity to the HER-2/neu protein after active immunization with HER-2/neu peptide-based vaccines. J Clin Oncol 2002; 20:2624 - 32; http://dx.doi.org/10.1200/JCO.2002.06.171; PMID: 12039923
  • Odunsi K, Qian F, Matsuzaki J, Mhawech-Fauceglia P, Andrews C, Hoffman EW, et al. 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 U S A 2007; 104:12837 - 42; http://dx.doi.org/10.1073/pnas.0703342104; PMID: 17652518
  • Diefenbach CS, Gnjatic S, Sabbatini P, Aghajanian C, Hensley ML, Spriggs DR, et al. Safety and immunogenicity study of NY-ESO-1b peptide and montanide ISA-51 vaccination of patients with epithelial ovarian cancer in high-risk first remission. Clin Cancer Res 2008; 14:2740 - 8; http://dx.doi.org/10.1158/1078-0432.CCR-07-4619; PMID: 18451240
  • Rahma OE, Ashtar E, Czystowska M, Szajnik ME, Wieckowski E, Bernstein S, et al. A gynecologic oncology group phase II trial of two p53 peptide vaccine approaches: subcutaneous injection and intravenous pulsed dendritic cells in high recurrence risk ovarian cancer patients. Cancer Immunol Immunother 2012; 61:373 - 84; http://dx.doi.org/10.1007/s00262-011-1100-9; PMID: 21927947
  • Leffers N, Lambeck AJ, Gooden MJ, Hoogeboom BN, Wolf R, Hamming IE, et al. Immunization with a P53 synthetic long peptide vaccine induces P53-specific immune responses in ovarian cancer patients, a phase II trial. Int J Cancer 2009; 125:2104 - 13; http://dx.doi.org/10.1002/ijc.24597; PMID: 19621448
  • Leffers N, Vermeij R, Hoogeboom BN, Schulze UR, Wolf R, Hamming IE, et al. Long-term clinical and immunological effects of p53-SLP® vaccine in patients with ovarian cancer. Int J Cancer 2012; 130:105 - 12; http://dx.doi.org/10.1002/ijc.25980; PMID: 21328579
  • Dohi S, Ohno S, Ohno Y, Takakura M, Kyo S, Soma G, et al. WT1 peptide vaccine stabilized intractable ovarian cancer patient for one year: a case report. Anticancer Res 2011; 31:2441 - 5; PMID: 21873157
  • Ohno S, Kyo S, Myojo S, Dohi S, Ishizaki J, Miyamoto K, et al. Wilms’ tumor 1 (WT1) peptide immunotherapy for gynecological malignancy. Anticancer Res 2009; 29:4779 - 84; PMID: 20032435
  • Sandmaier BM, Oparin DV, Holmberg LA, Reddish MA, MacLean GD, Longenecker BM. Evidence of a cellular immune response against sialyl-Tn in breast and ovarian cancer patients after high-dose chemotherapy, stem cell rescue, and immunization with Theratope STn-KLH cancer vaccine. J Immunother 1999; 22:54 - 66; http://dx.doi.org/10.1097/00002371-199901000-00008; PMID: 9924700
  • MacLean GD, Reddish MA, Koganty RR, Longenecker BM. Antibodies against mucin-associated sialyl-Tn epitopes correlate with survival of metastatic adenocarcinoma patients undergoing active specific immunotherapy with synthetic STn vaccine. J Immunother Emphasis Tumor Immunol 1996; 19:59 - 68; http://dx.doi.org/10.1097/00002371-199601000-00007; PMID: 8859725
  • Leffers N, Daemen T, Boezen HM, Melief KJ, Nijman HW. Vaccine-based clinical trials in ovarian cancer. Expert Rev Vaccines 2011; 10:775 - 84; http://dx.doi.org/10.1586/erv.11.42; PMID: 21692699
  • Odunsi K, Jungbluth AA, Stockert E, Qian F, Gnjatic S, Tammela J, et al. NY-ESO-1 and LAGE-1 cancer-testis antigens are potential targets for immunotherapy in epithelial ovarian cancer. Cancer Res 2003; 63:6076 - 83; PMID: 14522938
  • Vermeij R, Leffers N, Hoogeboom BN, Hamming IL, Wolf R, Reyners AK, et al. Potentiation of a p53-SLP vaccine by cyclophosphamide in ovarian cancer: A single-arm phase II study. Int J Cancer 2011; In press PMID: 22139992
  • Wang B, Kaumaya PT, Cohn DE. Immunization with synthetic VEGF peptides in ovarian cancer. Gynecol Oncol 2010; 119:564 - 70; http://dx.doi.org/10.1016/j.ygyno.2010.07.037; PMID: 20822802
  • Morse MA, Secord AA, Blackwell K, Hobeika AC, Sinnathamby G, Osada T, et al. MHC class I-presented tumor antigens identified in ovarian cancer by immunoproteomic analysis are targets for T-cell responses against breast and ovarian cancer. Clin Cancer Res 2011; 17:3408 - 19; http://dx.doi.org/10.1158/1078-0432.CCR-10-2614; PMID: 21300761
  • Chianese-Bullock KA, Irvin WP Jr., Petroni GR, Murphy C, Smolkin M, Olson WC, et al. A multipeptide vaccine is safe and elicits T-cell responses in participants with advanced stage ovarian cancer. J Immunother 2008; 31:420 - 30; http://dx.doi.org/10.1097/CJI.0b013e31816dad10; PMID: 18391753
  • Sabbatini PJ, Kudryashov V, Ragupathi G, Danishefsky SJ, Livingston PO, Bornmann W, et al. Immunization of ovarian cancer patients with a synthetic Lewis(y)-protein conjugate vaccine: a phase 1 trial. Int J Cancer 2000; 87:79 - 85; http://dx.doi.org/10.1002/1097-0215(20000701)87:1<79::AID-IJC12>3.0.CO;2-L; PMID: 10861456
  • Odunsi KRK. Lele S et al. Diversified prime and boost vaccination using recombinant vaccinia and fowlpox expressing NY-ESO-1 efficiently induces antibody, CD4+ and CD8+ anti tumor immune responses in patients with ovarian cancer. 38th Society of Gynecologic Oncologists Annual Meeting on Women's Cancer. San Diego, CA, USA, 2007.
  • Gulley JL, Arlen PM, Tsang KY, Yokokawa J, Palena C, Poole DJ, et al. Pilot study of vaccination with recombinant CEA-MUC-1-TRICOM poxviral-based vaccines in patients with metastatic carcinoma. Clin Cancer Res 2008; 14:3060 - 9; http://dx.doi.org/10.1158/1078-0432.CCR-08-0126; PMID: 18483372
  • Chiang CL, Kandalaft LE, Coukos G. Adjuvants for enhancing the immunogenicity of whole tumor cell vaccines. Int Rev Immunol 2011; 30:150 - 82; http://dx.doi.org/10.3109/08830185.2011.572210; PMID: 21557641
  • Buckanovich RJ, Facciabene A, Kim S, Benencia F, Sasaroli D, Balint K, et al. Endothelin B receptor mediates the endothelial barrier to T cell homing to tumors and disables immune therapy. Nat Med 2008; 14:28 - 36; http://dx.doi.org/10.1038/nm1699; PMID: 18157142
  • Ehlen TG, Hoskins PJ, Miller D, Whiteside TL, Nicodemus CF, Schultes BC, et al. A pilot phase 2 study of oregovomab murine monoclonal antibody to CA125 as an immunotherapeutic agent for recurrent ovarian cancer. Int J Gynecol Cancer 2005; 15:1023 - 34; http://dx.doi.org/10.1111/j.1525-1438.2005.00483.x; PMID: 16343178
  • Gordon AN, Schultes BC, Gallion H, Edwards R, Whiteside TL, Cermak JM, et al. CA125- and tumor-specific T-cell responses correlate with prolonged survival in oregovomab-treated recurrent ovarian cancer patients. Gynecol Oncol 2004; 94:340 - 51; http://dx.doi.org/10.1016/j.ygyno.2004.04.024; PMID: 15297171
  • Braly P, Nicodemus CF, Chu C, Collins Y, Edwards R, Gordon A, et al. The Immune adjuvant properties of front-line carboplatin-paclitaxel: a randomized phase 2 study of alternative schedules of intravenous oregovomab chemoimmunotherapy in advanced ovarian cancer. J Immunother 2009; 32:54 - 65; http://dx.doi.org/10.1097/CJI.0b013e31818b3dad; PMID: 19307994
  • Berek J, Taylor P, McGuire W, Smith LM, Schultes B, Nicodemus CF. Oregovomab maintenance monoimmunotherapy does not improve outcomes in advanced ovarian cancer. J Clin Oncol 2009; 27:418 - 25; http://dx.doi.org/10.1200/JCO.2008.17.8400; PMID: 19075271
  • Pfisterer J, du Bois A, Sehouli J, Loibl S, Reinartz S, Reuss A, et al. The anti-idiotypic antibody abagovomab in patients with recurrent ovarian cancer. A phase I trial of the AGO-OVAR. Ann Oncol 2006; 17:1568 - 77; http://dx.doi.org/10.1093/annonc/mdl357; PMID: 17005631
  • Reinartz S, Köhler S, Schlebusch H, Krista K, Giffels P, Renke K, et al. Vaccination of patients with advanced ovarian carcinoma with the anti-idiotype ACA125: immunological response and survival (phase Ib/II). Clin Cancer Res 2004; 10:1580 - 7; http://dx.doi.org/10.1158/1078-0432.CCR-03-0056; PMID: 15014007
  • Sabbatini P, Dupont J, Aghajanian C, Derosa F, Poynor E, Anderson S, et al. Phase I study of abagovomab in patients with epithelial ovarian, fallopian tube, or primary peritoneal cancer. Clin Cancer Res 2006; 12:5503 - 10; http://dx.doi.org/10.1158/1078-0432.CCR-05-2670; PMID: 17000686
  • Nicholson S, Bomphray CC, Thomas H, McIndoe A, Barton D, Gore M, et al. A phase I trial of idiotypic vaccination with HMFG1 in ovarian cancer. Cancer Immunol Immunother 2004; 53:809 - 16; http://dx.doi.org/10.1007/s00262-004-0522-z; PMID: 15127236
  • Hird V, Maraveyas A, Snook D, Dhokia B, Soutter WP, Meares C, et al. Adjuvant therapy of ovarian cancer with radioactive monoclonal antibody. Br J Cancer 1993; 68:403 - 6; http://dx.doi.org/10.1038/bjc.1993.349; PMID: 8347497
  • Oei AL, Verheijen RH, Seiden MV, Benigno BB, Lopes A, Soper JT, et al. Decreased intraperitoneal disease recurrence in epithelial ovarian cancer patients receiving intraperitoneal consolidation treatment with yttrium-90-labeled murine HMFG1 without improvement in overall survival. Int J Cancer 2007; 120:2710 - 4; http://dx.doi.org/10.1002/ijc.22663; PMID: 17354223
  • Bellone S, Siegel ER, Cocco E, Cargnelutti M, Silasi DA, Azodi M, et al. Overexpression of epithelial cell adhesion molecule in primary, metastatic, and recurrent/chemotherapy-resistant epithelial ovarian cancer: implications for epithelial cell adhesion molecule-specific immunotherapy. Int J Gynecol Cancer 2009; 19:860 - 6; http://dx.doi.org/10.1111/IGC.0b013e3181a8331f; PMID: 19574774
  • Heiss MM, Murawa P, Koralewski P, Kutarska E, Kolesnik OO, Ivanchenko VV, et al. The trifunctional antibody catumaxomab for the treatment of malignant ascites due to epithelial cancer: Results of a prospective randomized phase II/III trial. Int J Cancer 2010; 127:2209 - 21; http://dx.doi.org/10.1002/ijc.25423; PMID: 20473913
  • Baumann K, Pfisterer J, Wimberger P, Burchardi N, Kurzeder C, du Bois A, et al. Intraperitoneal treatment with the trifunctional bispecific antibody Catumaxomab in patients with platinum-resistant epithelial ovarian cancer: a phase IIa study of the AGO Study Group. Gynecol Oncol 2011; 123:27 - 32; http://dx.doi.org/10.1016/j.ygyno.2011.06.004; PMID: 21733566
  • Makhija S, Amler LC, Glenn D, Ueland FR, Gold MA, Dizon DS, et al. Clinical activity of gemcitabine plus pertuzumab in platinum-resistant ovarian cancer, fallopian tube cancer, or primary peritoneal cancer. J Clin Oncol 2010; 28:1215 - 23; http://dx.doi.org/10.1200/JCO.2009.22.3354; PMID: 19901115
  • Bookman MA, Darcy KM, Clarke-Pearson D, Boothby RA, Horowitz IR. Evaluation of monoclonal humanized anti-HER2 antibody, trastuzumab, in patients with recurrent or refractory ovarian or primary peritoneal carcinoma with overexpression of HER2: a phase II trial of the Gynecologic Oncology Group. J Clin Oncol 2003; 21:283 - 90; http://dx.doi.org/10.1200/JCO.2003.10.104; PMID: 12525520
  • Chung MK, Han SS, Roh JK. Synergistic embryotoxicity of combination pyrimethamine and folic acid in rats. Reprod Toxicol 1993; 7:463 - 8; http://dx.doi.org/10.1016/0890-6238(93)90091-K; PMID: 8274822
  • Molthoff CF, Prinssen HM, Kenemans P, van Hof AC, den Hollander W, Verheijen RH. Escalating protein doses of chimeric monoclonal antibody MOv18 immunoglobulin G in ovarian carcinoma patients: a phase I study. Cancer 1997; 80:Suppl 2712 - 20; http://dx.doi.org/10.1002/(SICI)1097-0142(19971215)80:12+<2712::AID-CNCR50>3.0.CO;2-B; PMID: 9406729
  • Konner JA, Bell-McGuinn KM, Sabbatini P, Hensley ML, Tew WP, Pandit-Taskar N, et al. Farletuzumab, a humanized monoclonal antibody against folate receptor alpha, in epithelial ovarian cancer: a phase I study. Clin Cancer Res 2010; 16:5288 - 95; http://dx.doi.org/10.1158/1078-0432.CCR-10-0700; PMID: 20855460
  • Perren TJ, Swart AM, Pfisterer J, Ledermann JA, Pujade-Lauraine E, Kristensen G, et al, ICON7 Investigators. A phase 3 trial of bevacizumab in ovarian cancer. N Engl J Med 2011; 365:2484 - 96; http://dx.doi.org/10.1056/NEJMoa1103799; PMID: 22204725
  • Burger RA, Brady MF, Bookman MA, Fleming GF, Monk BJ, Huang H, et al, Gynecologic Oncology Group. Incorporation of bevacizumab in the primary treatment of ovarian cancer. N Engl J Med 2011; 365:2473 - 83; http://dx.doi.org/10.1056/NEJMoa1104390; PMID: 22204724
  • Burger RA, Sill MW, Monk BJ, Greer BE, Sorosky JI. Phase II trial of bevacizumab in persistent or recurrent epithelial ovarian cancer or primary peritoneal cancer: a Gynecologic Oncology Group Study. J Clin Oncol 2007; 25:5165 - 71; http://dx.doi.org/10.1200/JCO.2007.11.5345; PMID: 18024863
  • Cannistra SA, Matulonis UA, Penson RT, Hambleton J, Dupont J, Mackey H, et al. Phase II study of bevacizumab in patients with platinum-resistant ovarian cancer or peritoneal serous cancer. J Clin Oncol 2007; 25:5180 - 6; http://dx.doi.org/10.1200/JCO.2007.12.0782; PMID: 18024865
  • Leffers N, Daemen T, Helfrich W, Boezen HM, Cohlen BJ, Melief K, et al. Antigen-specific active immunotherapy for ovarian cancer. Cochrane Database Syst Rev:CD007287
  • Rosenberg SA, Restifo NP, Yang JC, Morgan RA, Dudley ME. Adoptive cell transfer: a clinical path to effective cancer immunotherapy. Nat Rev Cancer 2008; 8:299 - 308; http://dx.doi.org/10.1038/nrc2355; PMID: 18354418
  • Dudley ME, Wunderlich JR, Yang JC, Sherry RM, Topalian SL, Restifo NP, et al. Adoptive cell transfer therapy following non-myeloablative but lymphodepleting chemotherapy for the treatment of patients with refractory metastatic melanoma. J Clin Oncol 2005; 23:2346 - 57; http://dx.doi.org/10.1200/JCO.2005.00.240; PMID: 15800326
  • Aoki Y, Takakuwa K, Kodama S, Tanaka K, Takahashi M, Tokunaga A, et al. Use of adoptive transfer of tumor-infiltrating lymphocytes alone or in combination with cisplatin-containing chemotherapy in patients with epithelial ovarian cancer. Cancer Res 1991; 51:1934 - 9; PMID: 2004379
  • Lum LG, LeFever AV, Treisman JS, Garlie NK, Hanson JP Jr.. Immune modulation in cancer patients after adoptive transfer of anti-CD3/anti-CD28-costimulated T cells-phase I clinical trial. J Immunother 2001; 24:408 - 19; http://dx.doi.org/10.1097/00002371-200109000-00003
  • Kershaw MH, Westwood JA, Parker LL, Wang G, Eshhar Z, Mavroukakis SA, et al. A phase I study on adoptive immunotherapy using gene-modified T cells for ovarian cancer. Clin Cancer Res 2006; 12:6106 - 15; http://dx.doi.org/10.1158/1078-0432.CCR-06-1183; PMID: 17062687
  • Geller MA, Cooley S, Judson PL, Ghebre R, Carson LF, Argenta PA, et al. A phase II study of allogeneic natural killer cell therapy to treat patients with recurrent ovarian and breast cancer. Cytotherapy 2011; 13:98 - 107; http://dx.doi.org/10.3109/14653249.2010.515582; PMID: 20849361
  • Steinman RM, Banchereau J. Taking dendritic cells into medicine. Nature 2007; 449:419 - 26; http://dx.doi.org/10.1038/nature06175; PMID: 17898760
  • Cannon MJ, O’Brien TJ. Cellular immunotherapy for ovarian cancer. Expert Opin Biol Ther 2009; 9:677 - 88; http://dx.doi.org/10.1517/14712590902932897; PMID: 19456205
  • Hernando JJ, Park TW, Kübler 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; http://dx.doi.org/10.1007/s00262-001-0255-1; PMID: 11845259
  • Chu CS, Boyer J, Schullery DS, Gimotty PA, Gamerman V, Bender J, et al. Phase I/II randomized trial of dendritic cell vaccination with or without cyclophosphamide for consolidation therapy of advanced ovarian cancer in first or second remission. Cancer Immunol Immunother 2012; 61:629 - 41; http://dx.doi.org/10.1007/s00262-011-1081-8; PMID: 22021066
  • Brossart P, Wirths S, Stuhler G, Reichardt VL, Kanz L, Brugger W. Induction of cytotoxic T-lymphocyte responses in vivo after vaccinations with peptide-pulsed dendritic cells. Blood 2000; 96:3102 - 8; PMID: 11049990
  • Hodi FS, Butler M, Oble DA, Seiden MV, Haluska FG, Kruse A, et al. Immunologic and clinical effects of antibody blockade of cytotoxic T lymphocyte-associated antigen 4 in previously vaccinated cancer patients. Proc Natl Acad Sci U S A 2008; 105:3005 - 10; http://dx.doi.org/10.1073/pnas.0712237105; PMID: 18287062
  • Berd D, Mastrangelo MJ. Effect of low dose cyclophosphamide on the immune system of cancer patients: reduction of T-suppressor function without depletion of the CD8+ subset. Cancer Res 1987; 47:3317 - 21; PMID: 2953413
  • 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 - 74; http://dx.doi.org/10.1084/jem.155.4.1063; PMID: 6460831
  • Ercolini AM, Ladle BH, Manning EA, Pfannenstiel LW, Armstrong TD, Machiels JP, et al. 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
  • Hodi FS, O’Day SJ, McDermott DF, Weber RW, Sosman JA, Haanen JB, et al. Improved survival with ipilimumab in patients with metastatic melanoma. N Engl J Med 2010; 363:711 - 23; http://dx.doi.org/10.1056/NEJMoa1003466; PMID: 20525992
  • Hodi FS, Mihm MC, Soiffer RJ, Haluska FG, Butler M, Seiden MV, et al. Biologic activity of cytotoxic T lymphocyte-associated antigen 4 antibody blockade in previously vaccinated metastatic melanoma and ovarian carcinoma patients. Proc Natl Acad Sci U S A 2003; 100:4712 - 7; http://dx.doi.org/10.1073/pnas.0830997100; PMID: 12682289
  • Alagkiozidis I, Facciabene A, Carpenito C, Benencia F, Jonak Z, Adams S, et al. Increased immunogenicity of surviving tumor cells enables cooperation between liposomal doxorubicin and IL-18. J Transl Med 2009; 7:104; http://dx.doi.org/10.1186/1479-5876-7-104; PMID: 20003308

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.