Skip to main content
NIHPA Author Manuscripts logoLink to NIHPA Author Manuscripts
. Author manuscript; available in PMC: 2017 Apr 1.
Published in final edited form as: Gynecol Oncol. 2016 Apr;141(1):86–94. doi: 10.1016/j.ygyno.2015.12.030

Leveraging Immunotherapy for the treatment of Gynecologic Cancers in the Era of Precision Medicine

Dmitriy Zamarin 1, Amir A Jazaeri 2,*
PMCID: PMC5007873  NIHMSID: NIHMS811467  PMID: 27016233

Abstract

During the past decade significant progress in the understanding of stimulatory and inhibitory signaling pathways in immune cells has reinvigorated the field of immuno-oncology. In this review we outline the current immunotherapy based approaches for the treatment of gynecological cancers, and focus on the emerging clinical data on immune checkpoint inhibitors, adoptive cell therapies, and vaccines. It is anticipated that in the coming years biomarker-guided clinical trials, will provide for a better understanding of the mechanisms of response and resistance to immunotherapy, and guide combination treatment strategies that will extend the benefit from immunotherapy to patients with gynecologic cancers.

Keywords: Immunotherapy, Ovarian Cancer, Cervical Cancer, Endometrial Cancer, Gynecologic cancers, Checkpoint Inhibitors, Immuno-oncology

Introduction

The immune system plays a key role in eliminating and controlling early tumor growth [1, 2]. Recognition and elimination of tumors by the immune system involves a series of steps coordinated by the various parts of the innate and adaptive immune system. The immune recognition of cancer begins at the tumor site, where fragments of malignant cells get taken up by professional antigen-presenting cells (APC) such as dendritic cells (DC). Activation of DCs in turn requires several maturation signals, which are in part provided by the “danger” signals released from the dying tumor cells, known as damage-associated molecular patterns (DAMPs) [3]. Following activation, APCs migrate to tumor-draining lymph nodes, where they present tumor-associated antigens (TAAs) in the form of antigenic peptides bound to the major histocompatibility complex (MHC) class I and II molecules. This enables antigen recognition by antigen-specific CD4 and CD8 T cells. In addition to recognition of specific antigenic peptides bound to MHC, activation of T cells requires another immunostimulatory signal, which is provided by engagement of a co-stimulatory receptor such as CD28 on the surface of T cells [4]. Activated T cells then migrate to tumors through the systemic vasculature by following a chemokine gradient [5, 6] and extravasate through a series of interaction with adhesion molecules in the tumor endothelium [7]. Finally, recognition of tumor targets proceeds through interaction between the T cell receptor (TCR) and specific antigenic tumor peptide bound to MHC, ultimately leading to T-cell mediated tumor destruction.

Starting with the process of antigen presentation, tumors have evolved a variety of resistance mechanisms that allow for successful escape from immune recognition and elimination [8]. Hence, immunotherapeutic approaches aim to improve recognition of tumors by the immune system and to inhibit the mechanisms of immune escape. Many of these approaches have been explored in gynecologic malignancies, with recent data demonstrating promising activity in various tumor types. Here we will discuss several examples of such modalities, primarily focusing on the more recently reported studies, though the list is certainly not exhaustive and multiple other approaches could be considered to be applicable. With emerging data, it is likely that a combination of several different modalities will be needed for optimal activation of anti-tumor immunity and therapeutic efficacy.

I. Immunotherapy of ovarian cancer

Epithelial ovarian cancer (EOC) is the fourth most common cancer in women and accounts for the highest number of gynecologic cancer deaths. Although EOC has not been traditionally considered a type of cancer that would be amenable to immunotherapy, multiple lines of evidence have demonstrated that the immune system likely plays a key role in eliminating and controlling ovarian cancer growth. In particular, the presence of tumor-infiltrating lymphocytes (TILs) has emerged as an important prognostic biomarker in EOC, with increased number of TILs predicting longer survival [9, 10]. Tumor-reactive antibodies and T cells have been demonstrated to be present in the peripheral blood of EOC patients [11, 12], and oligoclonal tumor-directed T cells have been directly isolated from the tumors and ascitic fluid [1320].

Based on these findings, several immunotherapeutic strategies have been explored in EOC. These approaches can be broadly subdivided into three categories: 1) Direct targeting of tumors with tumor-specific antibodies; 2) modalities that aim to enhance antigen presentation, such as vaccines, toll-like receptor (TLR) agonists, and oncolytic viruses, and 3) strategies focusing on activation of tumor-specific T cells, either through direct adoptive transfer or by targeting of activating and inhibitory pathways in T cells and tumor microenvironment.

A. Targeting of ovarian tumors with tumor-specific antibodies

Antibodies targeting surface antigens have been demonstrated to be effective against different cancer types [21, 22]. While some of these agents target tumor driver pathways (e.g. trastuzumab and cetuxumab), some in addition mediate antibody-dependent cellular cytotoxicity (ADCC), allowing for recognition of the antibody-labeled cancer cells with immune effectors, such as natural killer cells. In ovarian cancer, however, such strategies have been more elusive, likely secondary to lack of an optimal surface antigen. Indeed a 2014 Cochrane review of trials in ovarian cancer using antigen-specific targeting failed to establish a conclusive evidence for efficacy of such strategies in EOC [23].

CA-125

The extensive expression of CA125 and its cell-surface precursor MUC16 in the majority of ovarian carcinomas prompted several trials targeting CA125 [2426]. Oregovomab, an antibody targeting CA-125 has been evaluated in several studies, with early studies demonstrating the development of anti-CA-125 T cell responses [2729]. However, a randomized placebo-controlled phase III trial in patients with advanced EOC in first clinical remission failed to demonstrate benefit to oregovomab therapy, with no significant difference between the placebo and oregovomab groups [30].

EpCAM

Epithelial cell adhesion molecule (EpCAM) is a surface integrin receptor commonly overexpressed on cancer cells and appears to be associated with worsened prognosis in ovarian cancer [31]. Catumaxomab is a bispecific antibody recognizing EpCAM and T cell antigen CD3. In addition, catumaxomab possesses an ADCC-mediating Fc region, making it a trifunctional antibody. Catumaxomab mediates anti-tumor effect through two different immune mechanisms: via recruitment and activation of T cells to the EpCAM expressing tumor cells and via binding to Fc receptor-expressing effectors such as NK cells. In a phase II/III trial randomizing patients with advanced cancer with malignant ascites to standard paracentesis or paracentesis with intraperitoneal catumaxomab, catumaxomab delayed ascited re-accumulation, but had no impact on overall survival [32]. A variation on this strategy has been recently developed using bispecific T cell engagers (BiTE) recognizing EpCAM and CD3. This strategy has been demonstrated to be effective in xenograft models of human colorectal and ovarian cancer [33]. More recently, in preclinical models of ovarian carcinoma, a novel EpCAM-CD3 BiTE solitomab has demonstrated significant activity against human ovarian tumor cells in vitro and ex vivo [34, 35].

FRa

Folate receptor alpha is expressed in high frequency in epithelial ovarian cancer [36]. Farletuzumab, a monoclonal ADCC-mediating antibody against folate receptor alpha, was evaluated in several studies, with earlier studies demonstrating promising efficacy [37, 38]. Despite these findings, later larger studies in combination with chemotherapy in platinum-sensitive and resistant patients, however, failed to meet the primary endpoints (NCT00849667, NCT00738699). More recently, data from phase I study using IMGN853, a folate receptor alpha targeting antibody-drug conjugate in patients with FRa positive epithelial ovarian cancer and other Fra positive solid tumors demonstrated clinical benefit rate of 25–33% depending on schedule, with responses seen in different treatment schedule groups [39]. While this strategy is certainly promising, it is unclear whether there is any contribution of the immune system to the observed effect and further studies would be needed to answer this question.

B. Enhancement of recognition of tumor antigens by the immune system

Vaccines

Several different vaccination approaches have been explored in ovarian cancer [4044]. Those include simple vaccine preparations consisting of specific peptides and proteins, as well as more complex strategies, such as engineered cellular vaccines, DC vaccines, virus-vectored vaccines, and oncolytic viruses [4554]. A comprehensive review of different vaccination strategies that have been explored in ovarian cancer is published elsewhere [55]. The majority of the vaccines have focused on using cancer-testis antigens (e.g. NY-ESO-1), and proteins known to be overexpressed in EOC (e.g. p53, survivin, MUC1). In general, while the majority of the studies demonstrated evidence of cellular and antibody response to the antigens, clinical benefit afforded by vaccination has unfortunately been marginal at best. Since most of the strategies have relied on self-antigens, it is likely that vaccination alone is not sufficient to overcome the T cell tolerance and combinatorial therapies may be necessary. Indeed, studies in preclinical models indicate that combination of vaccines with immune checkpoint blockade result in enhancement over either approach alone [5663], thus generating rationale for exploration of similar strategies in human trials.

TLR agonists

Toll like receptors (TLR) are a class of proteins recognizing signature molecules that are broadly shared by various pathogens, and play a role in the innate immune response and tumor antigen processing and presentation by APC. Ligands for various TLRs are actively being explored as anti-cancer agents and there is a rationale for using such ligands in ovarian cancer [64]. VTX-2337 (motolimod) is a small molecule agonist of TLR8, which stimulates a strong innate immune response. VTX-2337 has been evaluated with systemic administration in combination with liposomal doxorubicin in animal models and in phase I study in patients with advanced ovarian cancer. The combination appeared to be safe, with evidence of immune activation and clinical benefit [65]. A phase 2 study evaluating motolimod in combination with liposomal doxorubicin is ongoing (NCT01666444). Another phase 1/2 study using combination of motolimod with liposomal doxorubicin and anti-PD-L1 antibody MEDI4736 is upcoming (NCT02431559).

Type I IFN

Type I IFN is an innate immune response cytokine, which plays a role in antiviral immune response. In addition, recent studies have demonstrated a critical role for the type I IFN pathway in anti-tumor immune response [66, 67], where type I IFN was demonstrated to be indispensible for tumor antigen cross-presentation by dendritic cells. Studies with systemic or intraperitoneal IFNα in patients with EOC failed to demonstrate significant efficacy, while often being associated with frequent dose-limiting toxicity [6870].

Oncolytic viruses

Although not initially thought of as immunotherapeutic agents, with evolving understanding of the interplay between oncolytic viruses and the immune system, came the recognition that virus-induced anti-tumor immune response, rather than direct tumor lysis, may be a dominant player driving the efficacy of these therapies. Several trials with intraperitoneally-administered oncolytic viruses have been conducted in ovarian cancer [7182]. In the majority of the studies, the treatment was well tolerated and while responses were rare, a good percentage of patients demonstrated stable disease, which was often durable. These studies suggest that intraperitoneal oncolytic viruses present a viable therapeutic strategy in ovarian cancer, though for optimal efficacy their evaluation in combination with other modalities (e.g. chemotherapy, other immunotherapies) is likely warranted.

C. Activation of tumor-specific T cells

Cytokines

IL-2 is a T-cell growth factor, which is FDA approved for treatment of renal cell cancer and malignant melanoma [83, 84]. IL-12 is a cytokine mainly produced by activated monocytes, tissue macrophages, and B cells. It can induce IFN-γ and together with IL-2 becomes a potent activator of cytotoxic T lymphocytes and NK cells [85, 86]. While both cytokines are associated with significant toxicity with systemic administration, the toxicity appears to be lower with locoregional (e.g. intraperitoneal) administration. A phase 1–2 study of IP IL-2 in patients with persistent or recurrent ovarian cancer had shown an overall response rate of 25.7%, with an overall 5-year survival probability of 13.9% [87]. As a different strategy, IL-12 expressing plasmid DNA was evaluated in patients with malignant melanoma with patients showing promising responses with intratumoral injection and electroporation [88, 89]. In ovarian cancer, an IP-administered IL-12-expressing plasmid was evaluated in combination with chemotherapy, and appeared to be well-tolerated, though responses did not exceed what would be expected with chemotherapy alone [90]. A subsequent study evaluated intraperitoneal EGEN-001, an IL-12 plasmid formulated with lipopolymer in patients with persistent or recurrent ovarian cancer. In this study of 22 patients, 35% had stable disease [91].

Immune checkpoint blockade

Activation of tumor-specific T cells requires binding of the T cell receptor to the tumor-specific antigen peptide presented by major histocompatibility complex (MHC) class I and II molecules on the surface of APC. In addition to recognition of cognate MHC-peptide complex, there is a requirement for another immunostimulatory signal, which is provided by activation of a co-stimulatory receptor such as CD28 on the surface of T cells [4]. In addition to CD28, T cells express a wide repertoire of other co-inhibitory and co-stimulatory receptors, which integrate a complex immune signaling network regulating T cell activation, differentiation, survival, and effector function [4]. Targeting of such receptors demonstrated significant activity in pre-clinical models and in early clinical trials [92]. The CTLA-4 and PD-1 inhibitory immune checkpoint blocking antibodies are the most advanced in clinical development and were recently approved by the FDA for treatment of metastatic melanoma [93, 94]. Based on these findings, therapy with immune checkpoint blockade (ICB) is being evaluated in preliminary trials in patients with EOC. A recent phase II study of Nivolumab in platinum resistant ovarian cancer patients showed response and disease control rates of 15 and 45%, respectively, including two patients with a durable complete response [95].

-CTLA-4

The CTLA-4- blocking antibody ipilimumab was the first immune checkpoint blocking antibody that has entered clinical testing and has an FDA-approved indication for treatment of metastatic melanoma on the basis of survival benefit demonstrated in a phase III study[93]. In eleven patients with ovarian cancer, who previously received autologous tumor cell vaccine expressing GM-CSF (GVAX), treatment with ipilimumab led to an objective response in one patient, which was durable for over 4 years [96].

-PD-1/PD-L1

A rationale for targeting PD-1/PD-L1 pathway in gynecologic malignancies was initially demonstrated in a phase I study of anti-PD-L1 antibody in patients with advanced cancer, which included 17 patients with ovarian cancer [97]. Of the ovarian cancer cohort, 22% of the patients had evidence of objective response or stable disease, lasting at least 24 weeks [97]. Preliminary clinical data have now in addition been reported for the EOC patients treated with PD-1 blocking drugs nivolumab and pembrolizumab, as well as PD-L1 blocking antibody avelumab. In a phase I study of nivolumab in 20 EOC patients, best overall response was 15%, including 2 patients with durable CR, with a total disease control rate of 45% [95]. In a phase Ib open-label expansion trial of avelumab in 75 patients with recurrent or refractory ovarian cancer, efficacy data from the first 23 patients demonstrated PR in 4 patients (17%), stable disease in 11 patients (48%), and 2 patients with PR after initial progression. In a phase I study of pembrolizumab in 26 ovarian cancer patients selected for PD-L1 positivity, the drug demonstrated activity with one patient with CR, 2 patients with PR, and 6 patients with stable disease. In all studies, the drugs were very well tolerated [95, 98, 99]. Based on these studies PD-1/PD-L1 blockade thus demonstrates promising activity in advanced ovarian cancer patients and larger studies are currently underway.

The development of CTLA-4 and PD-1/PD-L1-targeting antibodies has provided for an opportunity for evaluation of combinations of these agents, which resulted in additive or even synergistic activity in animal models of melanoma and ovarian cancer [100, 101] and in phase I dose escalation studies in metastatic melanoma and renal cell carcinoma [102, 103]. In melanoma, combined CTLA-4 and PD-1 blockade with ipilimumab and nivolumab, respectively, was recently demonstrated to enhance response rate and PFS in comparison to either agent alone, although with increased toxicity [104]. To determine whether addition of CTLA-4 blockade would increase therapeutic efficacy in EOC, there is an ongoing NRG Oncology Group randomized phase II study comparing the combination of nivolumab and ipilimumab to nivolumab alone in patients with relapsed EOC (NCT02498600).

D. Adoptive T cell therapies

Adoptive cell therapies (ACT) aim to overcome the immunosuppressive effect of tumor microenvironment through infusion of large numbers of autologous tumor-reactive T cells that have been expanded from tumor infiltrating lymphocytes (TILs) in vitro. Significant activity of such therapies have been reported in patients with metastatic melanoma [105]. Two trials have evaluated the efficacy of adoptive cell therapy in ovarian cancer with evidence of clinical benefit seen in the majority of the treated patients [106, 107]. These studies are obviously biased by selection of the patients from whom sufficient amount of TILs could be isolated, as patients with high numbers of TILs would be expected to have a more favorable prognosis. Additional studies using TIL ACT in ovarian cancer are ongoing (NCT02482090, NCT01883297).

Engineered T cells present an alternative strategy that avoids the need to isolate a sufficient number of TILs. Using this strategy, autologous lymphocytes isolated from peripheral blood are transduced either with a T cell receptor recognizing a specific tumor antigen within the context of MHC, or with a chimeric antigen receptor (CAR) recognizing a tumor-associated surface antigen [108]. Preclinical studies demonstrated that targeting of MUC16 with engineered T cells expressing a MUC16-specific CAR could induce complete eradication of orthotopic ovarian xenografts [109, 110], and a phase I study targeting MUC16 with CAR T cells is upcoming [111] (NCT02498912). It is conceivable that any antigens expressed on the surface of ovarian cancer could be targeted with these strategies, and studies are currently underway using T cells targeting differentiation proteins such as folate receptor alpha [112], and mesothelin (NCT01583686), and cancer testis antigens such as NY-ESO-1 (NCT01567891, NCT02457650).

While ACT, including engineered T cells, have demonstrated significant promise against several tumor types, they can be associated with significant toxicities, which include cytokine release syndrome (CRS) and toxicities from ‘on target, off tumor’ recognition [108]. CRS, resulting from activation of the infused tumor-specific T cells, can lead to a range of clinical toxicities, including fever, hypotension, hypoxia, and neurologic toxicities, and requires prompt recognition and treatment with steroids or anti-interleukin-6 receptor antibody tocilizumab [113, 114]. On target toxicities include recognition of normal tissues expressing the tumor antigen, and can lead to severe toxicities which have been fatal in some instances [115]. Engineering of additional safety features such as suicide genes into T cells may provide a safety switch and will likely be required in future studies exploring tumor antigen targets that are also expressed on normal tissues.

E. Other immunotherapeutic modalities on the horizon for ovarian cancer

Antibodies targeting co-stimulatory receptors

In addition to the antibodies targeting CTLA-4 and PD-1/PD-L1, antibodies to other T cell co-stimulatory and co-inhibitory receptors are currently in development. Amongst those are the agonistic antibodies targeting activating T-cell surface receptors (e.g. anti-GITR, anti-OX40) as well as other blocking antibodies to the inhibitory receptors on T-cells and NK cells (e.g. anti-LAG3, anti-KIR) (NCT01968109, NCT02061761, NCT01750580, NCT01714739, NCT01239134) [92]. Some of the studies are using combination of these drugs with PD-1/PD-L1 blocking antibodies, with the aim of reversal of T cell dysfunction or enhancement of T cell activation.

Targeting mechanisms of immunotherapy resistance

Several inhibitory mechanisms that play a role in immune evasion have been demonstrated to be associated with poor prognosis in ovarian cancer, including increased tumor-infiltrating regulatory T cells (Tregs) [10, 116, 117] and tumor-associated macrophages with an M2-immunophenotype (CD163+/CD68+) [118, 119], and expression of the immune inhibitory ligands such as PD-L1, B7-H3, and B7-H4, and of the inhibitory enzyme indoleamine 2,3-dioxygenase (IDO) by the tumor or stromal cells [120123]. Drugs targeting these mechanisms are currently being explored against solid tumors, including ovarian cancer.

Depletion or inhibition of the immunosuppressive cells in the tumor microenvironment has the potential to enhance the efficacy of immunotherapies. In animal models, combination of CTLA-4 blockade and depletion of regulatory T cells with anti-CD25 antibody resulted in improved therapeutic efficacy of CTLA-4 blockade [124]. Strategies targeting CD25 in humans with daclizumab [125] or denileukin diftitox (Ontak) [126] have also been evaluated, though benefit so far has been demonstrated to be marginal, likely because CD25 is not a marker specific for regulatory T cells and is also expressed on activated T effector lymphocytes. An anti-CCR4 antibody has been demonstrated to selectively deplete regulatory T cells from humans [127, 128], and a phase I study of the anti CCR4 antibody mogamulizumab in patients with solid tumors is currently ongoing (NCT01929486). In separate studies, mogamulizumab is being explored in combination with PD-L1 blocking antibody MEDI4736 and CTLA-4 blocking antibody tremelimumab (NCT02301130) and anti-PD-1 antibody nivolumab (NCT02476123).

In support of the immunosuppressive role of the myeloid cells in tumor microenvironment, study by Zhu et al. recently used a mouse model of pancreatic ductal adenocarcinoma to demonstrate that depletion of MDSC with CSF1 receptor antibody synergizes with PD-1 and CTLA-4 blockade [129]. A CSF1R- targeting antibody emactuzumab (RG7155) has been evaluated in patients with advanced solid tumors. Treatment with antibody led to depletion of tumor associated macrophages, with partial metabolic responses and disease stabilization seen in 5/44 and 6/40 patients respectively [130]. A phase Ib study of emactuzumab in combination with anti-PD-L1 antibody MPDL3280A in solid tumors including ovarian cancer is ongoing (NCT02323191). PLX3397, a small molecule inhibitor of CSF1R and c-kit is currently being evaluated in combination with paclitaxel (NCT01525602), as well as anti-PD-1 antibody pembrolizumab (NCT02452424) in patients with advanced solid tumors.

II. Immunotherapy for Endometrial Cancer

Endometrial cancer is the most common gynecologic malignancy in developed countries. While hysterectomy alone results in excellent cancer-related outcomes for patients with grade 1 and 2, low stage endometrioid tumors, the prognosis for patients with advanced stage disease and high risk histological subtypes remains poor. The normal human endometrium can be viewed as having the unique immunological roles of serving as a barrier to ascending infections from the female genital tract, and at the same time, harboring an immunosuppressive state that is crucial to gestation and fetal development. Despite this long recognized dual function our knowledge of the immune function of the endometrium and its alterations in malignant an premalignant states is remarkably incomplete [131].

Until recently, immunotherapy approaches used in the treatment of endometrial cancers have been largely limited to small series of patients with dendritic cell vaccines and related approaches [132134]. Not surprisingly, the recent success of immune checkpoint inhibitors in melanoma and other cancers has led to the investigation of these agents in endometrial cancers. Of particular interest is the fact approximately 20–30% of endometrial cancers are characterized by high microsatellite instability (MSI-H) due to genetic or epigenetic defects in components of the DNA mismatch repair pathway. These defects results in a high somatic mutation load and accordingly increased number of neoantigens in these MSI-H tumors [135, 136]. In colon cancer, MSI-H tumors have been shown to be more immunogenic with increased infiltration of immune cells and increased immune checkpoint expression[137, 138]. A recently published phase 2 study of pembrolizumab (an anti-PD-1 antibody) demonstrated object response rates of 40 and 71% in MSI-H colorectal and non-colorectal cohorts (which included 2 patients with endometrial cancer), respectively[139]. This same study found no responses in patients with microsatellite stable colorectal tumors (0 out of 18). Additionally, a third arm of the study was composed of non-colorectal MSI-H tumors, including two endometrial cancer cases. This group also showed improved objective response rate and progression free survival [139]. These results have generated great interest for clinical testing of immune checkpoint inhibitors in other MSI-H tumors including endometrial cancer.

Mutations in the replicative DNA polymerase epsilon (POLE) define another subset of highly immunogenic endometrial cancers characterized by ultra-high somatic mutations rates resulting from defects in the proof-reading function of this polymerase [136, 140, 141]. Approximately 5% of endometrial cancers are characterized by POLE mutations [142]. These tumor are predominantly endometrioid, grade III, and associated with peritumoral and tumor infiltrating lymphocytes [143]. POLE mutated tumors were reported to have the highest number of predicted neoantigens per tumor sample, followed by MSI-H tumors, and microsatellite stable tumors [136]. In addition, POLE and MSI-H tumors exhibited higher numbers of CD8+ TIL which were characterized by PD1 overexpression [136]. Taken together, these results provide a strong rationale for clinical investigations of checkpoint inhibitors and other immunotherapeutic approaches in endometrial cancers harboring the POLE ultramutated phenotype.

Other endometrial cancer immunotherapy targets currently under investigation include tissue factor (TF) [144], human trophoblast-cell-surface marker (Trop-2) [145], and survivin [146]. These targets are still in the preclinical or early clinical development with encouraging results.

III. Immunotherapy for cervical cancer and other HPV-related Gynecological Malignancies

Cervical cancer is unique among gynecologic malignant tumors because of its well-established and causative risk factor, chronic HPV infection. HPV, a double-stranded circular DNA virus, is the most common sexually transmitted infection, and it is estimated to infect 75–80% of women at one time or another during their lives [147]. The vast majority of HPV infections are cleared by the immune system, but a small fraction become chronic and result in cervical dysplasia and carcinoma. Over 170 HPV subtypes have been identified. However, HPV-16 and HPV-18 are the genotypes most commonly associated with cervical cancer, accounting for approximately 70% of invasive cervical cancers [148].

The infectious etiology of cervical cancer has led to the development of effective preventative vaccines. Discussion of preventative vaccines is beyond the scope of this review and interested readers are referred to excellent recent reviews [149, 150]. Despite the excellent potential for prevention, advanced stage/metastatic disease remain a principal cause of gynecologic cancer mortality in much of the world where there is limited access to vaccination and screening. Thus improved approaches for treatment of advanced cervical cancer including immunotherapy remain high clinical priorities.

Cervical cancer directed immunotherapies can be divided into several subtypes including therapeutic vaccines, immune checkpoint inhibitors, and adoptive cell therapies. These are briefly highlighted below.

A. Therapeutic vaccines

The success of preventative immunization has raised hopes for the successful development of a therapeutic vaccines. However, important differences between the two approach exist. To be effective prophylactic vaccines need only to block viral entry into cervical epithelial cells. In contrast, therapeutic vaccines must target integrated HPV virus that has become intracellular and is characterized by a non-lytic cycle. The main targets of therapeutic vaccine development have been HPV E6 and E7 proteins, since they are continuously expressed by infected cells and necessary maintenance of the malignant phenotype [151]. Targeting E6 and E7 has been accomplished using a number of different approaches including peptides, fusion proteins, and recombinant modified vaccinia and Listeria based vaccines [152, 153]. While a detailed discussion of these approaches is beyond the scope of this review, some of the main clinically relevant findings are briefly outlined below.

Vaccinia based vaccines

Modified vaccinia virus Ankara (MVA) is a double-stranded DNA virus derived from a Turkish smallpox vaccine strain. It is unable to replicate in most mammalian cells, has an excellent safety profile in humans, and can be genetically engineered to express foreign proteins [154]. MVA vaccines induce both humoral and cellular immune responses against the expressed foreign antigen. Vaccinia virus expressing the E6 and E7 proteins of HPV 16 and 18 has been developed and used for the treatment of HPV related intraepithelial neoplasia [155, 156]. In a study that included nearly 1200 women, subjects were injected with 107 virus particles directly into the dysplastic area (cervix, urethra, vulva, or anus). Complete elimination of the lesion was observed in 89.3% of patients and 83% had undetectable HPV DNA after treatment [155].

Listeria based vaccines

Similar to the vaccinia approach, Listeria monocytogenes (Lm) based vaccines use a genetically modified non-pathogenic form of this bacterium to present specific proteins/antigens to the immune system. ADXS11-001, is a vaccine designed to induce an immune response against HPV E7 [157]. The efficacy of ADXS11-001 against HPV-related human cervical, oropharyngeal and anal cancers is under evaluation in several ongoing clinical trials [158].

Peptide and protein vaccines

Peptide based-vaccines targeting HPV E6 and E7 [159] and non-HPV tumor associated antigens [160] are undergoing clinical testing. An alternative approach is the use of a Mycobacterium bovis heat shock protein (HSP65) linked to the entire HPV16 E7 sequence [161, 162]. Studies on high grade cervical intraepithelial neoplasia (CIN) have shown some complete responses, but due to the single arm design, the results are confounded by the potential for spontaneous regression [161, 162]. Another investigation using HPV16 E6 and E7 synthetic long peptide vaccine reported a complete clinical response in 47% of patients with VIN III [163].

Other vaccine approaches

Investigations using a number of other vaccine approaches targeting cervical cancer have been reported. These include genetically modified CEA expressing yeast, “naked DNA” constructs encoding E6–E7 [164166] and dendritic cell vaccines [167169]. However, most of these are either in early clinical development or have yet to demonstrate significant clinical responses.

B. Immune checkpoint inhibitors

Recent studies have provided support for a potential role for immune checkpoint inhibitors in the development and spread of cervical cancer. PDL1 expression was observed in 95% of CIN and 80% of squamous cell carcinomas, but undetectable in normal cervical epithelial cells [170]. Furthermore, lymph nodes harboring metastatic cervical cancer were found to be characterized by high levels of PDL1+ APCs and FOXP3+ regulatory T cells (Tregs) [171]. The PD-1: PD-L1 pathway has also been implicated in mediating immune-resistance in HPV-associated head and neck squamous cell cancers [172]. Hence, there is accumulating evidence suggesting a potential therapeutic benefit for checkpoint inhibitors in cervical and other HPV related cancers.

In cervical cancer, CTLA4 directed therapy with ipilimumab after chemoradiation is in Phase I clinical trial evaluation (NCT01711515) in patients with locally advanced or metastatic cervical cancer. In addition several other trials using agents targeting the PDL1-PD1 pathway (including a phase II GOG/NRG sponsored study (NCT02257528)) are targeting patients with advanced or recurrent cervical and head and neck cancers (clinicaltrials.gov).

C. Adoptive T cell therapies

HPV-reactive T cells can be be isolated and expanded from patients’ peripheral blood and tumor tissue [173, 174]. Stevanovic and colleagues recently showed that adoptive cell therapy using tumor infiltrating lymphocytes (TIL) resulted in two partial and one complete responses among the nine patients with heavily treated recurrent metastatic cervical cancer included in this cases series [173]. Of note, HPV reactivity of TIL correlated positively with clinical response, although the small number of cases precludes any definitive conclusions.

Adoptive cell therapy is one of the most rapidly evolving fields among immunotherapies, and there is significant research aimed at designing antigen specific engineered T cell receptors (TCRs) and chimeric antigen receptors (CARs). An NCI sponsored trial of adoptive cell therapy using an E6 targeting TCR (NCT02280811) is currently accruing.

IV. Immunotherapy of other Gynecologic Cancers

There is a relative paucity of data on potential immunotherapeutic targets in rarer gynecologic cancers such as sarcomas, GTN, low grade serous ovarian cancer, and malignant sex-cord stromal ovarian tumors. However, the basic immunotherapy concepts of enhancing immune recognition and activation and blocking immune-inhibitory pathways are likely to apply to these tumors as well.

Conclusions

The advances in understanding of genetics, tumor microenvironment, and interaction of tumors with the immune system in gynecologic malignancies provide compelling evidence that these cancers are not immunologically inert and generate a strong rationale for immunotherapeutic approaches. While many of such approaches have demonstrated significant promise in different tumor types, including gynecologic cancers, the benefit afforded by these treatments has so far been limited to only a subset of patients. Such challenges in immunotherapy logically call for identification of targetable markers predicting better response and development of rational combinatorial approaches. Data from preclinical studies indicate that combinatorial modalities targeting different parts of the immune response (e.g. vaccines and immune checkpoint blockade) result in improved therapeutic efficacy and early clinical studies indeed indicate that such approaches can indeed be more effective. Combined immune checkpoint blockade such as CTLA-4/PD-1 blockade has already demonstrated evidence of superior activity in metastatic melanoma and certainly warrants evaluation in gynecologic cancers. One must take these findings with a word of caution, however, as the side effect profile in patients with advanced gynecologic cancers may be different and might not justify the potential benefits. Furthermore, the significant toxicities reported from the combination regimens such combined CTLA-4/PD-1 blockade would make it challenging to build further treatment combinations based on this platform [104]. To address these problems it is becoming increasingly evident that the efficacy of specific therapies and combinations will likely not be universal and that the choice of a treatment modality may need to be tailored to fit the needs of each individual patient. Through biomarker-guided clinical trials, we’ll be able to better understand the mechanisms of response and resistance to immunotherapy and develop treatment strategies that will extend the benefit from immunotherapy to a broader range of patients and tumor types.

Acknowledgments

D.Z. received funding from Foundation for Women’s Cancers (Judith Liebenthal Robinson Ovarian Cancer Foundation Award). The authors would like acknowledge colleagues whose important contributions to the field of gynecologic cancer immunology was not cited due to space limitations.

Footnotes

Conflicts of Interest

The authors report no relevant conflicts of interest.

References

  • 1.Koebel CM, Vermi W, Swann JB, Zerafa N, Rodig SJ, Old LJ, et al. Adaptive immunity maintains occult cancer in an equilibrium state. Nature. 2007;450:903–7. doi: 10.1038/nature06309. [DOI] [PubMed] [Google Scholar]
  • 2.Willimsky G, Blankenstein T. Sporadic immunogenic tumours avoid destruction by inducing T-cell tolerance. Nature. 2005;437:141–6. doi: 10.1038/nature03954. [DOI] [PubMed] [Google Scholar]
  • 3.Kono H, Rock KL. How dying cells alert the immune system to danger. Nat Rev Immunol. 2008;8:279–89. doi: 10.1038/nri2215. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 4.Chen L, Flies DB. Molecular mechanisms of T cell co-stimulation and co-inhibition. Nat Rev Immunol. 2013;13:227–42. doi: 10.1038/nri3405. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 5.Harlin H, Meng Y, Peterson AC, Zha Y, Tretiakova M, Slingluff C, et al. Chemokine expression in melanoma metastases associated with CD8+ T-cell recruitment. Cancer research. 2009;69:3077–85. doi: 10.1158/0008-5472.CAN-08-2281. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 6.Franciszkiewicz K, Boissonnas A, Boutet M, Combadiere C, Mami-Chouaib F. Role of chemokines and chemokine receptors in shaping the effector phase of the antitumor immune response. Cancer Res. 2012;72:6325–32. doi: 10.1158/0008-5472.CAN-12-2027. [DOI] [PubMed] [Google Scholar]
  • 7.Zitvogel L, Tesniere A, Kroemer G. Cancer despite immunosurveillance: immunoselection and immunosubversion. Nat Rev Immunol. 2006;6:715–27. doi: 10.1038/nri1936. [DOI] [PubMed] [Google Scholar]
  • 8.Motz GT, Coukos G. Deciphering and reversing tumor immune suppression. Immunity. 2013;39:61–73. doi: 10.1016/j.immuni.2013.07.005. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 9.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. The New England journal of medicine. 2003;348:203–13. doi: 10.1056/NEJMoa020177. [DOI] [PubMed] [Google Scholar]
  • 10.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. Proceedings of the National Academy of Sciences of the United States of America. 2005;102:18538–43. doi: 10.1073/pnas.0509182102. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 11.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. Clinical cancer research : an official journal of the American Association for Cancer Research. 2003;9:1517–27. [PubMed] [Google Scholar]
  • 12.Goodell V, Salazar LG, Urban N, Drescher CW, Gray H, Swensen RE, et al. Antibody immunity to the p53 oncogenic protein is a prognostic indicator in ovarian cancer. Journal of clinical oncology : official journal of the American Society of Clinical Oncology. 2006;24:762–8. doi: 10.1200/JCO.2005.03.2813. [DOI] [PubMed] [Google Scholar]
  • 13.Hayashi K, Yonamine K, Masuko-Hongo K, Iida T, Yamamoto K, Nishioka K, et al. Clonal expansion of T cells that are specific for autologous ovarian tumor among tumor-infiltrating T cells in humans. Gynecologic oncology. 1999;74:86–92. doi: 10.1006/gyno.1999.5430. [DOI] [PubMed] [Google Scholar]
  • 14.Halapi E, Yamamoto Y, Juhlin C, Jeddi-Tehrani M, Grunewald J, Andersson R, et al. Restricted T cell receptor V-beta and J-beta usage in T cells from interleukin-2-cultured lymphocytes of ovarian and renal carcinomas. Cancer immunology, immunotherapy : CII. 1993;36:191–7. doi: 10.1007/BF01741091. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 15.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. The Journal of experimental medicine. 1995;181:2109–17. doi: 10.1084/jem.181.6.2109. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 16.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;12:429–37. [PubMed] [Google Scholar]
  • 17.Peoples GE, Schoof DD, Andrews JV, Goedegebuure PS, Eberlein TJ. T-cell recognition of ovarian cancer. Surgery. 1993;114:227–34. [PubMed] [Google Scholar]
  • 18.Dadmarz RD, Ordoubadi A, Mixon A, Thompson CO, Barracchini KC, Hijazi YM, et al. Tumor-infiltrating lymphocytes from human ovarian cancer patients recognize autologous tumor in an MHC class II-restricted fashion. Cancer J Sci Am. 1996;2:263–72. [PubMed] [Google Scholar]
  • 19.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. Obstetrics and gynecology. 2000;96:422–30. doi: 10.1016/s0029-7844(00)00916-9. [DOI] [PubMed] [Google Scholar]
  • 20.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. Proceedings of the National Academy of Sciences of the United States of America. 1995;92:432–6. doi: 10.1073/pnas.92.2.432. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 21.Bellati F, Napoletano C, Gasparri ML, Visconti V, Zizzari IG, Ruscito I, et al. Monoclonal antibodies in gynecological cancer: a critical point of view. Clinical & developmental immunology. 2011;2011:890758. doi: 10.1155/2011/890758. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 22.Reichert JM, Dhimolea E. The future of antibodies as cancer drugs. Drug discovery today. 2012 doi: 10.1016/j.drudis.2012.04.006. [DOI] [PubMed] [Google Scholar]
  • 23.Leffers N, Daemen T, Helfrich W, Boezen HM, Cohlen BJ, Melief CJ, et al. Antigen-specific active immunotherapy for ovarian cancer. The Cochrane database of systematic reviews. 2014;9:CD007287. doi: 10.1002/14651858.CD007287.pub3. [DOI] [PubMed] [Google Scholar]
  • 24.Bast RC, Jr, Feeney M, Lazarus H, Nadler LM, Colvin RB, Knapp RC. Reactivity of a monoclonal antibody with human ovarian carcinoma. The Journal of clinical investigation. 1981;68:1331–7. doi: 10.1172/JCI110380. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 25.Yin BW, Dnistrian A, Lloyd KO. Ovarian cancer antigen CA125 is encoded by the MUC16 mucin gene. International journal of cancer Journal international du cancer. 2002;98:737–40. doi: 10.1002/ijc.10250. [DOI] [PubMed] [Google Scholar]
  • 26.O’Brien TJ, Beard JB, Underwood LJ, Dennis RA, Santin AD, York L. The CA 125 gene: an extracellular superstructure dominated by repeat sequences. Tumour Biol. 2001;22:348–66. doi: 10.1159/000050638. [DOI] [PubMed] [Google Scholar]
  • 27.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. Gynecologic oncology. 2004;94:340–51. doi: 10.1016/j.ygyno.2004.04.024. [DOI] [PubMed] [Google Scholar]
  • 28.Mobus VJ, Baum RP, Bolle M, Kreienberg R, Noujaim AA, Schultes BC, et al. Immune responses to murine monoclonal antibody-B43.13 correlate with prolonged survival of women with recurrent ovarian cancer. Am J Obstet Gynecol. 2003;189:28–36. doi: 10.1067/mob.2003.347. [DOI] [PubMed] [Google Scholar]
  • 29.Noujaim AA, Schultes BC, Baum RP, Madiyalakan R. Induction of CA125-specific B and T cell responses in patients injected with MAb-B43.13--evidence for antibody-mediated antigen-processing and presentation of CA125 in vivo. Cancer biotherapy & radiopharmaceuticals. 2001;16:187–203. doi: 10.1089/10849780152389384. [DOI] [PubMed] [Google Scholar]
  • 30.Berek J, Taylor P, McGuire W, Smith LM, Schultes B, Nicodemus CF. Oregovomab maintenance monoimmunotherapy does not improve outcomes in advanced ovarian cancer. Journal of Clinical Oncology. 2009;27:418–25. doi: 10.1200/JCO.2008.17.8400. [DOI] [PubMed] [Google Scholar]
  • 31.Spizzo G, Went P, Dirnhofer S, Obrist P, Moch H, Baeuerle PA, et al. Overexpression of epithelial cell adhesion molecule (Ep-CAM) is an independent prognostic marker for reduced survival of patients with epithelial ovarian cancer. Gynecologic oncology. 2006;103:483–8. doi: 10.1016/j.ygyno.2006.03.035. [DOI] [PubMed] [Google Scholar]
  • 32.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. International journal of cancer Journal international du cancer. 2010;127:2209–21. doi: 10.1002/ijc.25423. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 33.Schlereth B, Fichtner I, Lorenczewski G, Kleindienst P, Brischwein K, da Silva A, et al. Eradication of tumors from a human colon cancer cell line and from ovarian cancer metastases in immunodeficient mice by a single-chain Ep-CAM-/CD3-bispecific antibody construct. Cancer Res. 2005;65:2882–9. doi: 10.1158/0008-5472.CAN-04-2637. [DOI] [PubMed] [Google Scholar]
  • 34.Ferrari F, Bellone S, Black J, Schwab CL, Lopez S, Cocco E, et al. Solitomab, an EpCAM/CD3 bispecific antibody construct (BiTE(R)), is highly active against primary uterine and ovarian carcinosarcoma cell lines in vitro. Journal of experimental & clinical cancer research : CR. 2015;34:123. doi: 10.1186/s13046-015-0241-7. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 35.English DP, Bellone S, Schwab CL, Roque DM, Lopez S, Bortolomai I, et al. Solitomab, an epithelial cell adhesion molecule/CD3 bispecific antibody (BiTE), is highly active against primary chemotherapy-resistant ovarian cancer cell lines in vitro and fresh tumor cells ex vivo. Cancer. 2015;121:403–12. doi: 10.1002/cncr.29062. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 36.Ross JF, Chaudhuri PK, Ratnam M. Differential regulation of folate receptor isoforms in normal and malignant tissues in vivo and in established cell lines. Physiologic and clinical implications. Cancer. 1994;73:2432–43. doi: 10.1002/1097-0142(19940501)73:9<2432::aid-cncr2820730929>3.0.co;2-s. [DOI] [PubMed] [Google Scholar]
  • 37.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. Clinical cancer research : an official journal of the American Association for Cancer Research. 2010;16:5288–95. doi: 10.1158/1078-0432.CCR-10-0700. [DOI] [PubMed] [Google Scholar]
  • 38.Armstrong DK, White AJ, Weil SC, Phillips M, Coleman RL. Farletuzumab (a monoclonal antibody against folate receptor alpha) in relapsed platinum-sensitive ovarian cancer. Gynecologic oncology. 2013;129:452–8. doi: 10.1016/j.ygyno.2013.03.002. [DOI] [PubMed] [Google Scholar]
  • 39.Borghaei H, O’Malley DM, Seward SM, Bauer TM, Perez RP, Oza AM, et al. Phase 1 study of IMGN853, a folate receptor alpha (FRα)-targeting antibody-drug conjugate (ADC) in patients (Pts) with epithelial ovarian cancer (EOC) and other FRA-positive solid tumors. J Clin Oncol. 2015;33(suppl) abstr 5558. [Google Scholar]
  • 40.Ninke Leffers TD, Helfrich Wijnand, Marike Boezen H, Cohlen Ben J, Melief Kees, Nijman Hans W. Antigen-specific active immunotherapy for ovarian cancer. doi: 10.1002/14651858.CD007287.pub2. unpublished. [DOI] [PubMed] [Google Scholar]
  • 41.Chu CS, Kim SH, June CH, Coukos G. Immunotherapy opportunities in ovarian cancer. Expert review of anticancer therapy. 2008;8:243–57. doi: 10.1586/14737140.8.2.243. [DOI] [PubMed] [Google Scholar]
  • 42.Odunsi K, Sabbatini P. Harnessing the immune system for ovarian cancer therapy. American journal of reproductive immunology (New York, NY : 1989) 2008;59:62–74. doi: 10.1111/j.1600-0897.2007.00560.x. [DOI] [PubMed] [Google Scholar]
  • 43.Sabbatini P, Odunsi K. Immunologic approaches to ovarian cancer treatment. Journal of clinical oncology : official journal of the American Society of Clinical Oncology. 2007;25:2884–93. doi: 10.1200/JCO.2007.11.0775. [DOI] [PubMed] [Google Scholar]
  • 44.Hung CF, Wu TC, Monie A, Roden R. Antigen-specific immunotherapy of cervical and ovarian cancer. Immunol Rev. 2008;222:43–69. doi: 10.1111/j.1600-065X.2008.00622.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 45.Sabbatini P, Tsuji T, Ferran L, Ritter E, Sedrak C, Tuballes K, et al. Phase I trial of overlapping long peptides from a tumor self-antigen and poly-ICLC shows rapid induction of integrated immune response in ovarian cancer patients. Clinical cancer research : an official journal of the American Association for Cancer Research. 2012;18:6497–508. doi: 10.1158/1078-0432.CCR-12-2189. [DOI] [PubMed] [Google Scholar]
  • 46.Sabbatini PJ, Ragupathi G, Hood C, Aghajanian CA, Juretzka M, Iasonos A, et al. Pilot study of a heptavalent vaccine-keyhole limpet hemocyanin conjugate plus QS21 in patients with epithelial ovarian, fallopian tube, or peritoneal cancer. Clinical cancer research : an official journal of the American Association for Cancer Research. 2007;13:4170–7. doi: 10.1158/1078-0432.CCR-06-2949. [DOI] [PubMed] [Google Scholar]
  • 47.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. International journal of cancer Journal international du cancer. 2009;125:2104–13. doi: 10.1002/ijc.24597. [DOI] [PubMed] [Google Scholar]
  • 48.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. Journal of immunotherapy (Hagerstown, Md : 1997) 2008;31:420–30. doi: 10.1097/CJI.0b013e31816dad10. [DOI] [PubMed] [Google Scholar]
  • 49.Reinartz S, Kohler 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) Clinical cancer research : an official journal of the American Association for Cancer Research. 2004;10:1580–7. doi: 10.1158/1078-0432.ccr-03-0056. [DOI] [PubMed] [Google Scholar]
  • 50.Wilson M, Villella JA, Berinstein NL, Brown J, Lheureux S, Haley A, et al. Phase I/Ib clinical and immunologic assessment of immunotherapeutic vaccine, DPX-Survivac in women with ovarian, Fallopian tube, or peritoneal cancer (OC) J Clin Oncol. 2014;32(5s) suppl abstr 5555. [Google Scholar]
  • 51.Odunsi K, Matsuzaki J, Karbach J, Neumann A, Mhawech-Fauceglia P, Miller A, et al. Efficacy of vaccination with recombinant vaccinia and fowlpox vectors expressing NY-ESO-1 antigen in ovarian cancer and melanoma patients. Proceedings of the National Academy of Sciences of the United States of America. 2012;109:5797–802. doi: 10.1073/pnas.1117208109. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 52.Mohebtash M, Tsang KY, Madan RA, Huen NY, Poole DJ, Jochems C, et al. A pilot study of MUC-1/CEA/TRICOM poxviral-based vaccine in patients with metastatic breast and ovarian cancer. Clinical cancer research : an official journal of the American Association for Cancer Research. 2011;17:7164–73. doi: 10.1158/1078-0432.CCR-11-0649. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 53.Gray HJ, Gargosky SE. Progression-free survival in ovarian cancer patients in second remission with mucin-1 autologous dendritic cell therapy. J Clin Oncol. 2014;32(5s) suppl abstr 5504. [Google Scholar]
  • 54.Sabbatini P, Berek J, Casado A, Cwiertka K, Pinter T, Pluzanska A, et al. Abagovomab maintenance therapy in patients with epithelial ovarian cancer after complete response post first line chemotherapy: preliminary results of the randomized double blind, placebo controlled multi-center MIMOSA trial. J Clin Onc. 2010:28. [Google Scholar]
  • 55.Liao JB, Disis ML. Therapeutic vaccines for ovarian cancer. Gynecologic oncology. 2013;130:667–73. doi: 10.1016/j.ygyno.2013.06.023. [DOI] [PubMed] [Google Scholar]
  • 56.van Elsas A, Hurwitz AA, Allison JP. Combination immunotherapy of B16 melanoma using anti-cytotoxic T lymphocyte-associated antigen 4 (CTLA-4) and granulocyte/macrophage colony-stimulating factor (GM-CSF)-producing vaccines induces rejection of subcutaneous and metastatic tumors accompanied by autoimmune depigmentation. The Journal of experimental medicine. 1999;190:355–66. doi: 10.1084/jem.190.3.355. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 57.Hurwitz AA, Yu TF, Leach DR, Allison JP. CTLA-4 blockade synergizes with tumor-derived granulocyte-macrophage colony-stimulating factor for treatment of an experimental mammary carcinoma. Proceedings of the National Academy of Sciences of the United States of America. 1998;95:10067–71. doi: 10.1073/pnas.95.17.10067. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 58.Curran MA, Allison JP. Tumor vaccines expressing flt3 ligand synergize with ctla-4 blockade to reject preimplanted tumors. Cancer research. 2009;69:7747–55. doi: 10.1158/0008-5472.CAN-08-3289. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 59.Wilcox RA, Flies DB, Zhu G, Johnson AJ, Tamada K, Chapoval AI, et al. Provision of antigen and CD137 signaling breaks immunological ignorance, promoting regression of poorly immunogenic tumors. The Journal of clinical investigation. 2002;109:651–9. doi: 10.1172/JCI14184. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 60.Ito D, Ogasawara K, Iwabuchi K, Inuyama Y, Onoe K. Induction of CTL responses by simultaneous administration of liposomal peptide vaccine with anti-CD40 and anti-CTLA-4 mAb. Journal of immunology. 2000;164:1230–5. doi: 10.4049/jimmunol.164.3.1230. [DOI] [PubMed] [Google Scholar]
  • 61.Saha A, Chatterjee SK. Combination of CTL-associated antigen-4 blockade and depletion of CD25 regulatory T cells enhance tumour immunity of dendritic cell-based vaccine in a mouse model of colon cancer. Scand J Immunol. 2010;71:70–82. doi: 10.1111/j.1365-3083.2009.02355.x. [DOI] [PubMed] [Google Scholar]
  • 62.Pedersen AE, Buus S, Claesson MH. Treatment of transplanted CT26 tumour with dendritic cell vaccine in combination with blockade of vascular endothelial growth factor receptor 2 and CTLA-4. Cancer letters. 2006;235:229–38. doi: 10.1016/j.canlet.2005.04.012. [DOI] [PubMed] [Google Scholar]
  • 63.Met O, Wang M, Pedersen AE, Nissen MH, Buus S, Claesson MH. The effect of a therapeutic dendritic cell-based cancer vaccination depends on the blockage of CTLA-4 signaling. Cancer letters. 2006;231:247–56. doi: 10.1016/j.canlet.2005.02.005. [DOI] [PubMed] [Google Scholar]
  • 64.Muccioli M, Benencia F. Toll-like Receptors in Ovarian Cancer as Targets for Immunotherapies. Frontiers in immunology. 2014;5:341. doi: 10.3389/fimmu.2014.00341. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 65.Monk BJ, Brady WE, Lankes HA, Facciabene A, Manjarrez K, Hershberg RM, et al. VTX-2337, a TLR8 agonist, plus chemotherapy in recurrent ovarian cancer: Preclinical and phase I data by the Gynecologic Oncology Group. J Clin Oncol. 2013;31(suppl) abstr 3077. [Google Scholar]
  • 66.Fuertes MB, Kacha AK, Kline J, Woo SR, Kranz DM, Murphy KM, et al. Host type I IFN signals are required for antitumor CD8+ T cell responses through CD8{alpha}+ dendritic cells. The Journal of experimental medicine. 2011;208:2005–16. doi: 10.1084/jem.20101159. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 67.Diamond MS, Kinder M, Matsushita H, Mashayekhi M, Dunn GP, Archambault JM, et al. Type I interferon is selectively required by dendritic cells for immune rejection of tumors. The Journal of experimental medicine. 2011;208:1989–2003. doi: 10.1084/jem.20101158. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 68.Abdulhay G, DiSaia PJ, Blessing JA, Creasman WT. Human lymphoblastoid interferon in the treatment of advanced epithelial ovarian malignancies: a Gynecologic Oncology Group Study. Am J Obstet Gynecol. 1985;152:418–23. doi: 10.1016/s0002-9378(85)80151-4. [DOI] [PubMed] [Google Scholar]
  • 69.Hall GD, Brown JM, Coleman RE, Stead M, Metcalf KS, Peel KR, et al. Maintenance treatment with interferon for advanced ovarian cancer: results of the Northern and Yorkshire gynaecology group randomised phase III study. British journal of cancer. 2004;91:621–6. doi: 10.1038/sj.bjc.6602037. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 70.Alberts DS, Hannigan EV, Liu PY, Jiang C, Wilczynski S, Copeland L, et al. Randomized trial of adjuvant intraperitoneal alpha-interferon in stage III ovarian cancer patients who have no evidence of disease after primary surgery and chemotherapy: An intergroup study. Gynecologic oncology. 2006;100:133–8. doi: 10.1016/j.ygyno.2005.07.117. [DOI] [PubMed] [Google Scholar]
  • 71.Kim KH, Dmitriev IP, Saddekni S, Kashentseva EA, Harris RD, Aurigemma R, et al. A phase I clinical trial of Ad5/3-Delta24, a novel serotype-chimeric, infectivity-enhanced, conditionally-replicative adenovirus (CRAd), in patients with recurrent ovarian cancer. Gynecologic oncology. 2013;130:518–24. doi: 10.1016/j.ygyno.2013.06.003. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 72.Kim KH, Dmitriev I, O’Malley JP, Wang M, Saddekni S, You Z, et al. A phase I clinical trial of Ad5.SSTR/TK.RGD, a novel infectivity-enhanced bicistronic adenovirus, in patients with recurrent gynecologic cancer. Clinical cancer research : an official journal of the American Association for Cancer Research. 2012;18:3440–51. doi: 10.1158/1078-0432.CCR-11-2852. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 73.Kimball KJ, Preuss MA, Barnes MN, Wang M, Siegal GP, Wan W, et al. A phase I study of a tropism-modified conditionally replicative adenovirus for recurrent malignant gynecologic diseases. Clinical cancer research : an official journal of the American Association for Cancer Research. 2010;16:5277–87. doi: 10.1158/1078-0432.CCR-10-0791. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 74.Galanis E, Hartmann LC, Cliby WA, Long HJ, Peethambaram PP, Barrette BA, et al. Phase I trial of intraperitoneal administration of an oncolytic measles virus strain engineered to express carcinoembryonic antigen for recurrent ovarian cancer. Cancer research. 2010;70:875–82. doi: 10.1158/0008-5472.CAN-09-2762. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 75.Wolf JK, Bodurka DC, Gano JB, Deavers M, Ramondetta L, Ramirez PT, et al. A phase I study of Adp53 (INGN 201; ADVEXIN) for patients with platinum- and paclitaxel-resistant epithelial ovarian cancer. Gynecologic oncology. 2004;94:442–8. doi: 10.1016/j.ygyno.2004.05.041. [DOI] [PubMed] [Google Scholar]
  • 76.Vasey PA, Shulman LN, Campos S, Davis J, Gore M, Johnston S, et al. Phase I trial of intraperitoneal injection of the E1B-55-kd-gene-deleted adenovirus ONYX-015 (dl1520) given on days 1 through 5 every 3 weeks in patients with recurrent/refractory epithelial ovarian cancer. Journal of clinical oncology : official journal of the American Society of Clinical Oncology. 2002;20:1562–9. doi: 10.1200/JCO.2002.20.6.1562. [DOI] [PubMed] [Google Scholar]
  • 77.Hemminki A, Wang M, Desmond RA, Strong TV, Alvarez RD, Curiel DT. Serum and ascites neutralizing antibodies in ovarian cancer patients treated with intraperitoneal adenoviral gene therapy. Human gene therapy. 2002;13:1505–14. doi: 10.1089/10430340260185139. [DOI] [PubMed] [Google Scholar]
  • 78.Hasenburg A, Fischer DC, Tong XW, Rojas-Martinez A, Nyberg-Hoffman C, Orlowska-Volk M, et al. Histologic and immunohistochemical analysis of tissue response to adenovirus-mediated herpes simplex thymidine kinase gene therapy of ovarian cancer. International journal of gynecological cancer : official journal of the International Gynecological Cancer Society. 2002;12:66–73. doi: 10.1046/j.1525-1438.2002.01068.x. [DOI] [PubMed] [Google Scholar]
  • 79.Buller RE, Runnebaum IB, Karlan BY, Horowitz JA, Shahin M, Buekers T, et al. A phase I/II trial of rAd/p53 (SCH 58500) gene replacement in recurrent ovarian cancer. Cancer Gene Ther. 2002;9:553–66. doi: 10.1038/sj.cgt.7700472. [DOI] [PubMed] [Google Scholar]
  • 80.Hasenburg A, Tong XW, Fischer DC, Rojas-Martinez A, Nyberg-Hoffman C, Kaplan AL, et al. Adenovirus-mediated thymidine kinase gene therapy in combination with topotecan for patients with recurrent ovarian cancer: 2.5-year follow-up. Gynecologic oncology. 2001;83:549–54. doi: 10.1006/gyno.2001.6442. [DOI] [PubMed] [Google Scholar]
  • 81.Alvarez RD, Gomez-Navarro J, Wang M, Barnes MN, Strong TV, Arani RB, et al. Adenoviral-mediated suicide gene therapy for ovarian cancer. Molecular therapy : the journal of the American Society of Gene Therapy. 2000;2:524–30. doi: 10.1006/mthe.2000.0194. [DOI] [PubMed] [Google Scholar]
  • 82.Alvarez RD, Barnes MN, Gomez-Navarro J, Wang M, Strong TV, Arafat W, et al. A cancer gene therapy approach utilizing an anti-erbB-2 single-chain antibody-encoding adenovirus (AD21): a phase I trial. Clinical cancer research : an official journal of the American Association for Cancer Research. 2000;6:3081–7. [PubMed] [Google Scholar]
  • 83.Fyfe GA, Fisher RI, Rosenberg SA, Sznol M, Parkinson DR, Louie AC. Long-term response data for 255 patients with metastatic renal cell carcinoma treated with high-dose recombinant interleukin-2 therapy. Journal of Clinical Oncology. 1996;14:2410–1. doi: 10.1200/JCO.1996.14.8.2410. [DOI] [PubMed] [Google Scholar]
  • 84.Chang E, Rosenberg SA. Patients with melanoma metastases at cutaneous and subcutaneous sites are highly susceptible to interleukin-2-based therapy. Journal of immunotherapy (Hagerstown, Md : 1997) 2001;24:88–90. doi: 10.1097/00002371-200101000-00010. [DOI] [PubMed] [Google Scholar]
  • 85.Wolf SF, Temple PA, Kobayashi M, Young D, Dicig M, Lowe L, et al. Cloning of cDNA for natural killer cell stimulatory factor, a heterodimeric cytokine with multiple biologic effects on T and natural killer cells. Journal of immunology (Baltimore, Md : 1950) 1991;146:3074–81. [PubMed] [Google Scholar]
  • 86.Trinchieri G. Interleukin-12: a cytokine produced by antigen-presenting cells with immunoregulatory functions in the generation of T-helper cells type 1 and cytotoxic lymphocytes. Blood. 1994;84:4008–27. [PubMed] [Google Scholar]
  • 87.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. Journal of Clinical Oncology. 1997;15:3399–407. doi: 10.1200/JCO.1997.15.11.3399. [DOI] [PubMed] [Google Scholar]
  • 88.Mahvi DM, Henry MB, Albertini MR, Weber S, Meredith K, Schalch H, et al. Intratumoral injection of IL-12 plasmid DNA--results of a phase I/IB clinical trial. Cancer Gene Ther. 2007;14:717–23. doi: 10.1038/sj.cgt.7701064. [DOI] [PubMed] [Google Scholar]
  • 89.Daud AI, DeConti RC, Andrews S, Urbas P, Riker AI, Sondak VK, et al. Phase I trial of interleukin-12 plasmid electroporation in patients with metastatic melanoma. Journal of Clinical Oncology. 2008;26:5896–903. doi: 10.1200/JCO.2007.15.6794. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 90.Anwer K, Kelly FJ, Chu C, Fewell JG, Lewis D, Alvarez RD. Phase I trial of a formulated IL-12 plasmid in combination with carboplatin and docetaxel chemotherapy in the treatment of platinum-sensitive recurrent ovarian cancer. Gynecologic oncology. 2013;131:169–73. doi: 10.1016/j.ygyno.2013.07.081. [DOI] [PubMed] [Google Scholar]
  • 91.Alvarez RD, Sill MW, Davidson SA, Muller CY, Bender DP, DeBernardo RL, et al. A phase II trial of intraperitoneal EGEN-001, an IL-12 plasmid formulated with PEG-PEI-cholesterol lipopolymer in the treatment of persistent or recurrent epithelial ovarian, fallopian tube or primary peritoneal cancer: a gynecologic oncology group study. Gynecologic oncology. 2014;133:433–8. doi: 10.1016/j.ygyno.2014.03.571. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 92.Zamarin D, Postow MA. Immune checkpoint modulation: rational design of combination strategies. Pharmacology & therapeutics. 2015;150:23–32. doi: 10.1016/j.pharmthera.2015.01.003. [DOI] [PubMed] [Google Scholar]
  • 93.Hodi FS, O’Day SJ, McDermott DF, Weber RW, Sosman JA, Haanen JB, et al. Improved survival with ipilimumab in patients with metastatic melanoma. The New England journal of medicine. 2010;363:711–23. doi: 10.1056/NEJMoa1003466. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 94.Hamid O, Robert C, Daud A, Hodi FS, Hwu WJ, Kefford R, et al. Safety and tumor responses with lambrolizumab (anti-PD-1) in melanoma. The New England journal of medicine. 2013;369:134–44. doi: 10.1056/NEJMoa1305133. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 95.Hamanishi J, Mandai M, Ikeda T, Minami M, Kawaguchi A, Murayama T, et al. Safety and Antitumor Activity of Anti-PD-1 Antibody, Nivolumab, in Patients With Platinum-Resistant Ovarian Cancer. Journal of clinical oncology : official journal of the American Society of Clinical Oncology. 2015 doi: 10.1200/JCO.2015.62.3397. [DOI] [PubMed] [Google Scholar]
  • 96.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. Proceedings of the National Academy of Sciences of the United States of America. 2008;105:3005–10. doi: 10.1073/pnas.0712237105. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 97.Brahmer JR, Tykodi SS, Chow LQ, Hwu WJ, Topalian SL, Hwu P, et al. Safety and activity of anti-PD-L1 antibody in patients with advanced cancer. The New England journal of medicine. 2012;366:2455–65. doi: 10.1056/NEJMoa1200694. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 98.Disis ML, Patel MR, Pant S, Infante JR, Lockhart AC, Kelly K, et al. Avelumab (MSB0010718C), an anti-PD-L1 antibody, in patients with previously treated, recurrent or refractory ovarian cancer: A phase Ib, open-label expansion trial. Journal of clinical oncology : official journal of the American Society of Clinical Oncology. 2015;33(suppl) abstr 5509. [Google Scholar]
  • 99.AV, Piha-Paul SA, Ott PA, Mehnert JM, Berton-Rigaud D, Johnson EA, et al. Antitumor activity and safety of pembrolizumab in patients (pts) with PD-L1 positive advanced ovarian cancer: Interim results from a phase Ib study. Journal of clinical oncology : official journal of the American Society of Clinical Oncology. 2015;33(suppl) abstr 5510. [Google Scholar]
  • 100.Curran MA, Montalvo W, Yagita H, Allison JP. PD-1 and CTLA-4 combination blockade expands infiltrating T cells and reduces regulatory T and myeloid cells within B16 melanoma tumors. Proceedings of the National Academy of Sciences of the United States of America. 2010;107:4275–80. doi: 10.1073/pnas.0915174107. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 101.Duraiswamy J, Kaluza KM, Freeman GJ, Coukos G. Dual blockade of PD-1 and CTLA-4 combined with tumor vaccine effectively restores T-cell rejection function in tumors. Cancer Res. 2013;73:3591–603. doi: 10.1158/0008-5472.CAN-12-4100. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 102.Wolchok JD, Kluger H, Callahan MK, Postow MA, Rizvi NA, Lesokhin AM. Nivolumab plus Ipilimumab in Advanced Melanoma. New Engl J Med. 2013;369:122–33. doi: 10.1056/NEJMoa1302369. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 103.Hammers HJ, Plimack ER, Infante JR, Ernstoff MS, Rini BI, McDermott DF, et al. Phase I study of nivolumab in combination with ipilimumab in metastatic renal cell carcinoma (mRCC) Journal of Clinical Oncology. 2014;32(suppl) doi: 10.1200/JCO.2016.72.1985. abstr 4504. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 104.Larkin J, Chiarion-Sileni V, Gonzalez R, Grob JJ, Cowey CL, Lao CD, et al. Combined Nivolumab and Ipilimumab or Monotherapy in Untreated Melanoma. The New England journal of medicine. 2015 doi: 10.1056/NEJMoa1504030. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 105.Rosenberg SA, Packard BS, Aebersold PM, Solomon D, Topalian SL, Toy ST, et al. Use of tumor-infiltrating lymphocytes and interleukin-2 in the immunotherapy of patients with metastatic melanoma. A preliminary report. The New England journal of medicine. 1988;319:1676–80. doi: 10.1056/NEJM198812223192527. [DOI] [PubMed] [Google Scholar]
  • 106.Fujita K, Ikarashi H, Takakuwa K, Kodama S, Tokunaga A, Takahashi T, et al. Prolonged disease-free period in patients with advanced epithelial ovarian cancer after adoptive transfer of tumor-infiltrating lymphocytes. Clinical cancer research : an official journal of the American Association for Cancer Research. 1995;1:501–7. [PubMed] [Google Scholar]
  • 107.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 research. 1991;51:1934–9. [PubMed] [Google Scholar]
  • 108.Smith EL, Zamarin D, Lesokhin AM. Harnessing the immune system for cancer therapy. Current opinion in oncology. 2014;26:600–7. doi: 10.1097/CCO.0000000000000128. [DOI] [PubMed] [Google Scholar]
  • 109.Koneru M, Purdon TJ, Spriggs D, Koneru S, Brentjens RJ. IL-12 secreting tumor-targeted chimeric antigen receptor T cells eradicate ovarian tumors. Oncoimmunology. 2015;4:e994446. doi: 10.4161/2162402X.2014.994446. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 110.Chekmasova AA, Rao TD, Nikhamin Y, Park KJ, Levine DA, Spriggs DR, et al. Successful eradication of established peritoneal ovarian tumors in SCID-Beige mice following adoptive transfer of T cells genetically targeted to the MUC16 antigen. Clinical cancer research : an official journal of the American Association for Cancer Research. 2010;16:3594–606. doi: 10.1158/1078-0432.CCR-10-0192. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 111.Koneru M, O’Cearbhaill R, Pendharkar S, Spriggs DR, Brentjens RJ. A phase I clinical trial of adoptive T cell therapy using IL-12 secreting MUC-16(ecto) directed chimeric antigen receptors for recurrent ovarian cancer. Journal of translational medicine. 2015;13:102. doi: 10.1186/s12967-015-0460-x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 112.Kandalaft LE, Powell DJ, Jr, Coukos G. A phase I clinical trial of adoptive transfer of folate receptor-alpha redirected autologous T cells for recurrent ovarian cancer. Journal of translational medicine. 2012;10:157. doi: 10.1186/1479-5876-10-157. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 113.Davila ML, Riviere I, Wang X, Bartido S, Park J, Curran K, et al. Efficacy and toxicity management of 19-28z CAR T cell therapy in B cell acute lymphoblastic leukemia. Sci Transl Med. 2014;6:224ra25. doi: 10.1126/scitranslmed.3008226. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 114.Grupp SA, Kalos M, Barrett D, Aplenc R, Porter DL, Rheingold SR, et al. Chimeric antigen receptor-modified T cells for acute lymphoid leukemia. The New England journal of medicine. 2013;368:1509–18. doi: 10.1056/NEJMoa1215134. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 115.Morgan RA, Yang JC, Kitano M, Dudley ME, Laurencot CM, Rosenberg SA. Case report of a serious adverse event following the administration of T cells transduced with a chimeric antigen receptor recognizing ERBB2. Molecular therapy : the journal of the American Society of Gene Therapy. 2010;18:843–51. doi: 10.1038/mt.2010.24. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 116.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 research. 2001;61:4766–72. [PubMed] [Google Scholar]
  • 117.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. Nature medicine. 2004;10:942–9. doi: 10.1038/nm1093. [DOI] [PubMed] [Google Scholar]
  • 118.Lan C, Huang X, Lin S, Huang H, Cai Q, Wan T, et al. Expression of M2-polarized macrophages is associated with poor prognosis for advanced epithelial ovarian cancer. Technol Cancer Res Treat. 2013;12:259–67. doi: 10.7785/tcrt.2012.500312. [DOI] [PubMed] [Google Scholar]
  • 119.Reinartz S, Schumann T, Finkernagel F, Wortmann A, Jansen JM, Meissner W, et al. Mixed-polarization phenotype of ascites-associated macrophages in human ovarian carcinoma: correlation of CD163 expression, cytokine levels and early relapse. International journal of cancer Journal international du cancer. 2014;134:32–42. doi: 10.1002/ijc.28335. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 120.Kryczek I, Wei S, Zhu G, Myers L, Mottram P, Cheng P, et al. Relationship between B7-H4, regulatory T cells, and patient outcome in human ovarian carcinoma. Cancer research. 2007;67:8900–5. doi: 10.1158/0008-5472.CAN-07-1866. [DOI] [PubMed] [Google Scholar]
  • 121.Hamanishi J, Mandai M, Iwasaki M, Okazaki T, Tanaka Y, Yamaguchi K, et al. Programmed cell death 1 ligand 1 and tumor-infiltrating CD8+ T lymphocytes are prognostic factors of human ovarian cancer. Proceedings of the National Academy of Sciences of the United States of America. 2007;104:3360–5. doi: 10.1073/pnas.0611533104. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 122.Zang X, Sullivan PS, Soslow RA, Waitz R, Reuter VE, Wilton A, et al. Tumor associated endothelial expression of B7-H3 predicts survival in ovarian carcinomas. Mod Pathol. 2010;23:1104–12. doi: 10.1038/modpathol.2010.95. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 123.Inaba T, Ino K, Kajiyama H, Yamamoto E, Shibata K, Nawa A, et al. Role of the immunosuppressive enzyme indoleamine 2,3-dioxygenase in the progression of ovarian carcinoma. Gynecologic oncology. 2009;115:185–92. doi: 10.1016/j.ygyno.2009.07.015. [DOI] [PubMed] [Google Scholar]
  • 124.Sutmuller RP, van Duivenvoorde LM, van Elsas A, Schumacher TN, Wildenberg ME, Allison JP, et al. Synergism of cytotoxic T lymphocyte-associated antigen 4 blockade and depletion of CD25(+) regulatory T cells in antitumor therapy reveals alternative pathways for suppression of autoreactive cytotoxic T lymphocyte responses. The Journal of experimental medicine. 2001;194:823–32. doi: 10.1084/jem.194.6.823. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 125.Rech AJ, Vonderheide RH. Clinical use of anti-CD25 antibody daclizumab to enhance immune responses to tumor antigen vaccination by targeting regulatory T cells. Annals of the New York Academy of Sciences. 2009;1174:99–106. doi: 10.1111/j.1749-6632.2009.04939.x. [DOI] [PubMed] [Google Scholar]
  • 126.Telang S, Rasku MA, Clem AL, Carter K, Klarer AC, Badger WR, et al. Phase II trial of the regulatory T cell-depleting agent, denileukin diftitox, in patients with unresectable stage IV melanoma. BMC cancer. 2011;11:515. doi: 10.1186/1471-2407-11-515. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 127.Sugiyama D, Nishikawa H, Maeda Y, Nishioka M, Tanemura A, Katayama I, et al. Anti-CCR4 mAb selectively depletes effector-type FoxP3+CD4+ regulatory T cells, evoking antitumor immune responses in humans. Proceedings of the National Academy of Sciences of the United States of America. 2013;110:17945–50. doi: 10.1073/pnas.1316796110. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 128.Ogura M, Ishida T, Hatake K, Taniwaki M, Ando K, Tobinai K, et al. Multicenter phase II study of mogamulizumab (KW-0761), a defucosylated anti-cc chemokine receptor 4 antibody, in patients with relapsed peripheral T-cell lymphoma and cutaneous T-cell lymphoma. Journal of clinical oncology : official journal of the American Society of Clinical Oncology. 2014;32:1157–63. doi: 10.1200/JCO.2013.52.0924. [DOI] [PubMed] [Google Scholar]
  • 129.Zhu Y, Knolhoff BL, Meyer MA, Nywening TM, West BL, Luo J, et al. CSF1/CSF1R Blockade Reprograms Tumor-Infiltrating Macrophages and Improves Response to T Cell Checkpoint Immunotherapy in Pancreatic Cancer Models. Cancer research. 2014 doi: 10.1158/0008-5472.CAN-13-3723. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 130.Gomez-Roca CA, Cassier PA, Italiano A, Cannarile M, Ries C, Brillouet CM, et al. Phase I study of RG7155, a novel anti-CSF1R antibody, in patients with advanced/metastatic solid tumors. J Clin Oncol. 2015;33(suppl) abstr 3005. [Google Scholar]
  • 131.Vanderstraeten A, Tuyaerts S, Amant F. The immune system in the normal endometrium and implications for endometrial cancer development. Journal of reproductive immunology. 2015;109:7–16. doi: 10.1016/j.jri.2014.12.006. [DOI] [PubMed] [Google Scholar]
  • 132.Santin AD, Hermonat PL, Ravaggi A, Bellone S, Cowan C, Coke C, et al. Development and therapeutic effect of adoptively transferred T cells primed by tumor lysate-pulsed autologous dendritic cells in a patient with metastatic endometrial cancer. Gynecologic and obstetric investigation. 2000;49:194–203. doi: 10.1159/000010246. [DOI] [PubMed] [Google Scholar]
  • 133.Santin AD, Bellone S, Ravaggi A, Roman JJ, Pecorelli S, Parham GP, et al. Induction of tumour-specific CD8(+) cytotoxic T lymphocytes by tumour lysate-pulsed autologous dendritic cells in patients with uterine serous papillary cancer. British journal of cancer. 2002;86:151–7. doi: 10.1038/sj.bjc.6600026. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 134.Coosemans A, Vanderstraeten A, Tuyaerts S, Verschuere T, Moerman P, Berneman ZN, et al. Wilms’ Tumor Gene 1 (WT1)--loaded dendritic cell immunotherapy in patients with uterine tumors: a phase I/II clinical trial. Anticancer research. 2013;33:5495–500. [PubMed] [Google Scholar]
  • 135.Alexandrov LB, Nik-Zainal S, Wedge DC, Aparicio SA, Behjati S, Biankin AV, et al. Signatures of mutational processes in human cancer. Nature. 2013;500:415–21. doi: 10.1038/nature12477. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 136.Howitt BE, Shukla SA, Sholl LM, Ritterhouse LL, Watkins JC, Rodig S, et al. Association of Polymerase e-Mutated and Microsatellite-Instable Endometrial Cancers With Neoantigen Load, Number of Tumor-Infiltrating Lymphocytes, and Expression of PD-1 and PD-L1. JAMA Oncol. 2015 doi: 10.1001/jamaoncol.2015.2151. [DOI] [PubMed] [Google Scholar]
  • 137.Dolcetti R, Viel A, Doglioni C, Russo A, Guidoboni M, Capozzi E, et al. High prevalence of activated intraepithelial cytotoxic T lymphocytes and increased neoplastic cell apoptosis in colorectal carcinomas with microsatellite instability. The American journal of pathology. 1999;154:1805–13. doi: 10.1016/S0002-9440(10)65436-3. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 138.Llosa NJ, Cruise M, Tam A, Wicks EC, Hechenbleikner EM, Taube JM, et al. The vigorous immune microenvironment of microsatellite instable colon cancer is balanced by multiple counter-inhibitory checkpoints. Cancer discovery. 2015;5:43–51. doi: 10.1158/2159-8290.CD-14-0863. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 139.Le DT, Uram JN, Wang H, Bartlett BR, Kemberling H, Eyring AD, et al. PD-1 Blockade in Tumors with Mismatch-Repair Deficiency. The New England journal of medicine. 2015;372:2509–20. doi: 10.1056/NEJMoa1500596. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 140.Cancer Genome Atlas Research N. Kandoth C, Schultz N, Cherniack AD, Akbani R, Liu Y, et al. Integrated genomic characterization of endometrial carcinoma. Nature. 2013;497:67–73. doi: 10.1038/nature12113. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 141.Bellone S, Centritto F, Black J, Schwab C, English D, Cocco E, et al. Polymerase epsilon (POLE) ultra-mutated tumors induce robust tumor-specific CD4+ T cell responses in endometrial cancer patients. Gynecologic oncology. 2015;138:11–7. doi: 10.1016/j.ygyno.2015.04.027. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 142.Billingsley CC, Cohn DE, Mutch DG, Stephens JA, Suarez AA, Goodfellow PJ. Polymerase varepsilon (POLE) mutations in endometrial cancer: clinical outcomes and implications for Lynch syndrome testing. Cancer. 2015;121:386–94. doi: 10.1002/cncr.29046. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 143.Bakhsh S, Kinloch M, Hoang LN, Soslow R, Kobel M, Lee CH, et al. Histopathological features of endometrial carcinomas associated with POLE mutations: implications for decisions about adjuvant therapy. Histopathology. 2015 doi: 10.1111/his.12878. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 144.Cocco E, Hu Z, Richter CE, Bellone S, Casagrande F, Bellone M, et al. hI-con1, a factor VII-IgGFc chimeric protein targeting tissue factor for immunotherapy of uterine serous papillary carcinoma. British journal of cancer. 2010;103:812–9. doi: 10.1038/sj.bjc.6605760. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 145.Varughese J, Cocco E, Bellone S, de Leon M, Bellone M, Todeschini P, et al. Uterine serous papillary carcinomas overexpress human trophoblast-cell-surface marker (Trop-2) and are highly sensitive to immunotherapy with hRS7, a humanized anti-Trop-2 monoclonal antibody. Cancer. 2011;117:3163–72. doi: 10.1002/cncr.25891. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 146.Vanderstraeten A, Everaert T, Van Bree R, Verbist G, Luyten C, Amant F, et al. In Vitro Validation of Survivin as Target Tumor-associated Antigen for Immunotherapy in Uterine Cancer. Journal of immunotherapy (Hagerstown, Md : 1997) 2015;38:239–49. doi: 10.1097/CJI.0000000000000085. [DOI] [PubMed] [Google Scholar]
  • 147.Satterwhite CL, Torrone E, Meites E, Dunne EF, Mahajan R, Ocfemia MC, et al. Sexually transmitted infections among US women and men: prevalence and incidence estimates, 2008. Sexually transmitted diseases. 2013;40:187–93. doi: 10.1097/OLQ.0b013e318286bb53. [DOI] [PubMed] [Google Scholar]
  • 148.Clifford GM, Smith JS, Plummer M, Munoz N, Franceschi S. Human papillomavirus types in invasive cervical cancer worldwide: a meta-analysis. British journal of cancer. 2003;88:63–73. doi: 10.1038/sj.bjc.6600688. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 149.Kash N, Lee MA, Kollipara R, Downing C, Guidry J, Tyring SK. Safety and Efficacy Data on Vaccines and Immunization to Human Papillomavirus. Journal of clinical medicine. 2015;4:614–33. doi: 10.3390/jcm4040614. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 150.Brotherton JM, Ogilvie GS. Current status of human papillomavirus vaccination. Current opinion in oncology. 2015;27:399–404. doi: 10.1097/CCO.0000000000000211. [DOI] [PubMed] [Google Scholar]
  • 151.zur Hausen H. Immortalization of human cells and their malignant conversion by high risk human papillomavirus genotypes. Seminars in cancer biology. 1999;9:405–11. doi: 10.1006/scbi.1999.0144. [DOI] [PubMed] [Google Scholar]
  • 152.McKee SJ, Bergot AS, Leggatt GR. Recent progress in vaccination against human papillomavirus-mediated cervical cancer. Reviews in medical virology. 2015;25(Suppl 1):54–71. doi: 10.1002/rmv.1824. [DOI] [PubMed] [Google Scholar]
  • 153.Ma B, Maraj B, Tran NP, Knoff J, Chen A, Alvarez RD, et al. Emerging human papillomavirus vaccines. Expert opinion on emerging drugs. 2012;17:469–92. doi: 10.1517/14728214.2012.744393. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 154.Remy-Ziller C, Germain C, Spindler A, Hoffmann C, Silvestre N, Rooke R, et al. Immunological characterization of a modified vaccinia virus Ankara vector expressing the human papillomavirus 16 E1 protein. Clinical and vaccine immunology : CVI. 2014;21:147–55. doi: 10.1128/CVI.00678-13. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 155.Rosales R, Lopez-Contreras M, Rosales C, Magallanes-Molina JR, Gonzalez-Vergara R, Arroyo-Cazarez JM, et al. Regression of human papillomavirus intraepithelial lesions is induced by MVA E2 therapeutic vaccine. Human gene therapy. 2014;25:1035–49. doi: 10.1089/hum.2014.024. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 156.Borysiewicz LK, Fiander A, Nimako M, Man S, Wilkinson GW, Westmoreland D, et al. A recombinant vaccinia virus encoding human papillomavirus types 16 and 18, E6 and E7 proteins as immunotherapy for cervical cancer. Lancet. 1996;347:1523–7. doi: 10.1016/s0140-6736(96)90674-1. [DOI] [PubMed] [Google Scholar]
  • 157.Wallecha A, French C, Petit R, Singh R, Amin A, Rothman J. Lm-LLO-Based Immunotherapies and HPV-Associated Disease. Journal of oncology. 2012;2012:542851. doi: 10.1155/2012/542851. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 158.Cory L, Chu C. ADXS-HPV: a therapeutic Listeria vaccination targeting cervical cancers expressing the HPV E7 antigen. Human vaccines & immunotherapeutics. 2014;10:3190–5. doi: 10.4161/hv.34378. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 159.de Vos van Steenwijk PJ, Ramwadhdoebe TH, Lowik MJ, van der Minne CE, Berends-van der Meer DM, Fathers LM, et al. A placebo-controlled randomized HPV16 synthetic long-peptide vaccination study in women with high-grade cervical squamous intraepithelial lesions. Cancer immunology, immunotherapy : CII. 2012;61:1485–92. doi: 10.1007/s00262-012-1292-7. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 160.Kawano K, Tsuda N, Waki K, Matsueda S, Hata Y, Ushijima K, et al. Personalized peptide vaccination for cervical cancer patients who have received prior platinum-based chemotherapy. Cancer science. 2015;106:1111–7. doi: 10.1111/cas.12729. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 161.Roman LD, Wilczynski S, Muderspach LI, Burnett AF, O’Meara A, Brinkman JA, et al. A phase II study of Hsp-7 (SGN-00101) in women with high-grade cervical intraepithelial neoplasia. Gynecologic oncology. 2007;106:558–66. doi: 10.1016/j.ygyno.2007.05.038. [DOI] [PubMed] [Google Scholar]
  • 162.Einstein MH, Kadish AS, Burk RD, Kim MY, Wadler S, Streicher H, et al. Heat shock fusion protein-based immunotherapy for treatment of cervical intraepithelial neoplasia III. Gynecologic oncology. 2007;106:453–60. doi: 10.1016/j.ygyno.2007.04.038. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 163.Kenter GG, Welters MJ, Valentijn AR, Lowik MJ, Berends-van der Meer DM, Vloon AP, et al. Vaccination against HPV-16 oncoproteins for vulvar intraepithelial neoplasia. The New England journal of medicine. 2009;361:1838–47. doi: 10.1056/NEJMoa0810097. [DOI] [PubMed] [Google Scholar]
  • 164.Trimble CL, Peng S, Kos F, Gravitt P, Viscidi R, Sugar E, et al. A phase I trial of a human papillomavirus DNA vaccine for HPV16+ cervical intraepithelial neoplasia 2/3. Clinical cancer research : an official journal of the American Association for Cancer Research. 2009;15:361–7. doi: 10.1158/1078-0432.CCR-08-1725. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 165.Bloy N, Buque A, Aranda F, Castoldi F, Eggermont A, Cremer I, et al. Trial watch: Naked and vectored DNA-based anticancer vaccines. Oncoimmunology. 2015;4:e1026531. doi: 10.1080/2162402X.2015.1026531. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 166.Santin AD, Bellone S, Gokden M, Cannon MJ, Parham GP. Vaccination with HPV-18 E7-pulsed dendritic cells in a patient with metastatic cervical cancer. The New England journal of medicine. 2002;346:1752–3. doi: 10.1056/NEJM200205303462219. [DOI] [PubMed] [Google Scholar]
  • 167.Santin AD, Bellone S, Palmieri M, Zanolini A, Ravaggi A, Siegel ER, et al. Human papillomavirus type 16 and 18 E7-pulsed dendritic cell vaccination of stage IB or IIA cervical cancer patients: a phase I escalating-dose trial. Journal of virology. 2008;82:1968–79. doi: 10.1128/JVI.02343-07. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 168.Muderspach L, Wilczynski S, Roman L, Bade L, Felix J, Small LA, et al. A phase I trial of a human papillomavirus (HPV) peptide vaccine for women with high-grade cervical and vulvar intraepithelial neoplasia who are HPV 16 positive. Clinical cancer research : an official journal of the American Association for Cancer Research. 2000;6:3406–16. [PubMed] [Google Scholar]
  • 169.Santin AD, Bellone S, Palmieri M, Ravaggi A, Romani C, Tassi R, et al. HPV16/18 E7-pulsed dendritic cell vaccination in cervical cancer patients with recurrent disease refractory to standard treatment modalities. Gynecologic oncology. 2006;100:469–78. doi: 10.1016/j.ygyno.2005.09.040. [DOI] [PubMed] [Google Scholar]
  • 170.Mezache L, Paniccia B, Nyinawabera A, Nuovo GJ. Enhanced expression of PD L1 in cervical intraepithelial neoplasia and cervical cancers. Mod Pathol. 2015 doi: 10.1038/modpathol.2015.108. [DOI] [PubMed] [Google Scholar]
  • 171.Heeren AM, Koster BD, Samuels S, Ferns DM, Chondronasiou D, Kenter GG, et al. High and interrelated rates of PD-L1+CD14+ antigen-presenting cells and regulatory T cells mark the microenvironment of metastatic lymph nodes from patients with cervical cancer. Cancer immunology research. 2015;3:48–58. doi: 10.1158/2326-6066.CIR-14-0149. [DOI] [PubMed] [Google Scholar]
  • 172.Lyford-Pike S, Peng S, Young GD, Taube JM, Westra WH, Akpeng B, et al. Evidence for a role of the PD-1:PD-L1 pathway in immune resistance of HPV-associated head and neck squamous cell carcinoma. Cancer research. 2013;73:1733–41. doi: 10.1158/0008-5472.CAN-12-2384. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 173.Stevanovic S, Draper LM, Langhan MM, Campbell TE, Kwong ML, Wunderlich JR, et al. Complete regression of metastatic cervical cancer after treatment with human papillomavirus-targeted tumor-infiltrating T cells. Journal of clinical oncology : official journal of the American Society of Clinical Oncology. 2015;33:1543–50. doi: 10.1200/JCO.2014.58.9093. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 174.Ramos CA, Narala N, Vyas GM, Leen AM, Gerdemann U, Sturgis EM, et al. Human papillomavirus type 16 E6/E7-specific cytotoxic T lymphocytes for adoptive immunotherapy of HPV-associated malignancies. Journal of immunotherapy (Hagerstown, Md : 1997) 2013;36:66–76. doi: 10.1097/CJI.0b013e318279652e. [DOI] [PMC free article] [PubMed] [Google Scholar]

RESOURCES