Open access peer-reviewed chapter

Immunotherapy in Gynecologic Cancers

Written By

Marcus Vetter and Viola Heinzelmann-Schwarz

Submitted: 26 May 2016 Reviewed: 25 January 2017 Published: 26 April 2017

DOI: 10.5772/67605

From the Edited Volume

Immunotherapy - Myths, Reality, Ideas, Future

Edited by Krassimir Metodiev

Chapter metrics overview

1,767 Chapter Downloads

View Full Metrics

Abstract

During the last years, significant progress in the understanding of signaling pathways of immune cells has revive the field of immune therapy for cancer. In this chapter, we explain the recent immunotherapy-based strategies for the treatment of gynecological cancers including cervical cancer, endometrial cancer, ovarian cancer, and vulvar cancer. This work will mainly focus on emerging clinical data on immune checkpoint inhibitors. But also data on adoptive T cell therapies and vaccines will be presented. It is anticipated that in future biomarker-guided randomized trials will provide better approaches in terms of response and resistance to immune therapy. The use of combination therapy for gynecological cancer might be one possible approach to overcome resistance.

Keywords

  • gynecologic cancers
  • ovarian cancer
  • endometrial cancer
  • cervical cancer
  • immune therapy
  • immune checkpoint inhibitors

1. Introduction

Gynecologic cancers include vulva, vaginal, cervical, endometrial, and ovarian/tubal/peritoneal cancers, the latter of which are still classified as one disease. As these organ-classified cancers have different characteristics, biology, therapies, and outcomes, during the past decade, approaches have been undertaken to subclassify them as to their heterogeneity and based on next-generation profiling. The main cornerstone of treatment for gynecologic cancer comprises in most cancers of surgical resection with different possibilities of adjuvant further therapy like chemo-, radio-, targeted, and, increasingly, immunotherapy.

In the United States, almost 90,000 women were diagnosed with gynecologic cancers in 2015 and over 29,000 will die from their disease [1]. Many women are cured with combined modalities, however, in ovarian cancer, for example, over 70% of cancers are diagnosed in advanced International Federation of Gynecology and Obstetrics (FIGO) stage III or IV, thus their five-year overall survival is only 30% [2].

Outcome in ovarian cancer in all stages is the worst of all gynecological cancers with a 10-year overall survival of 30%, followed by vaginal cancer with a 10-year overall survival of 35%. Cervical and vulvar cancers have a 10-year overall survival rate of 65%. Endometrial cancer has the best prognosis, with a 10-year survival rate of 80% [1].

Immunotherapy represents a new alternative and rational approach for the treatment of cancer, including gynecologic cancers [3, 4]. More than a decade ago, it was demonstrated for ovarian cancer that tumor-infiltrating lymphocytes (TILs) play an important role in tumor rejection and prognosis [5]. This was one of the first evidence that immune therapy might be beneficial in ovarian cancer patients. A meta-analysis confirmed the prognostic role of TILs for ovarian cancer patients [6]. Later it was also demonstrated that the ratio of different T cell subtypes plays an important role [7].

A major function of the immune system is to continually seek out and eliminate cancer cells as they arise in a process defined as cancer immunosurveillance [8]. This involves both innate and adaptive immune mechanisms that function complimentarily to promote tumor immunity. Most importantly is that antitumor immune responses can be induced by immunological agents. Various forms of immunotherapies are central components of treatment regimens for a number of malignancies [9]. To eliminate cancer cells by T cells is only one-step in a complex immunity cycle [10].

In general, there are three strategies to treat cancer with immunotherapeutic approaches:

  1. Increase tumor antigen presentation.

  2. Increase T-cell activity.

  3. Targeting the tumor environment (immune inhibitory mechanisms).

Strategies to increase tumor antigen presentation includes vaccinations, use of innate immune activators, oncolytic viruses, type I interferon, and toll-like receptor (TLR) agonists.

Especially for epithelial ovarian cancer (EOC), several vaccination approaches have been applied, e.g., cellular vaccines, dendritic cell (DC) vaccines, and virus-loaded vaccines. Several studies have used overexpressed proteins in EOC as a target, e.g., p53, surviving, and MUC1. Several studies have demonstrated immune response but clinical benefit rate was minimal in all of these studies. The vaccination approach is not used in clinical practice nowadays [11].

To increase T-cell activity, there are several approaches tested including cytokine therapies with IL-2 and IL-12, the use of checkpoint inhibitors, and adoptive T cell therapies [12, 13]. Rosenberg et al. demonstrated in 2015 an elegant new therapeutic approach by generating tumor-associated antigen-specific T cells via expression of T-cell receptor or chimeric antigen receptor (CAR) [14]. With this approach, CD19 targeting CAR therapy for acute lymphatic leukemia of the B cell lineage was applied with a very high remission rate of 90% [15]. In ovarian cancer, adoptive T-cell therapy might be also effective. For example, NY-ESO-1 is specifically expressed in cancer, 42% expression has been seen in ovarian cancer. This might be an important target for adoptive T-cell therapy [16].

To target the tumor environment, there are also several therapeutic approaches. It has been demonstrated that several immune inhibitory mechanisms are associated with poor prognosis in gynecological cancer and in particular, in ovarian cancer, e.g., tumor infiltrating regulatory T cells, tumor-associated macrophages, expression of indoleamine 2, 3-dioxygenase (IDO) by tumor stromal cells [17, 18]. To target the responsible pathways might be effective, especially in combination with newer programmed cell death ligand-1 (PDL-1) or its receptor programmed cell death protein-1 (PD-1) checkpoint inhibition [19, 20].

For gynecological cancer, in particular for ovarian cancer, there is still an unmet challenge in cell therapy for cancer. The selection of the right target antigen, which is tumor cell-specific and has a robust expression, seems to be very important.

Advertisement

2. Cervical cancer

Cervical cancer is the fourth most common female cancer worldwide, with estimated 528,000 new cases and 266,000 deaths in 2014 [21]. Infection with high risk types of human papillomavirus (HPV) is the most crucial risk factor [22]. Human papillomavirus types 16, 18, 31, 33, 35, 39, 45, 51, 52, 56, 58, 59, 68, 72, and 82 are associated with high risk of cervical cancer, whereas HPV types 6, 11, 40, 42, 43, 44, 54, 61, 70, 72, and 81 are considered to have low carcinogenic risk [23, 24].

Most common cases are diagnosed in less developed countries, where cervical cancer comprises nearly 15% of cancers in women. In Switzerland, with a small population of only eight million, the incidence is much lower, with about 240 cases diagnosed each year [25].

Better screening methods and vaccination against HPV in the past decades have led to an improvement of cervical cancer prognosis in developed countries, particularly where a broad prevention plan has been put in place [26]. To date, we have an efficacious vaccination available against the nine most important HPV types (HPV 6, 11, 16, 18, 31, 33, 45, 52, and 58) [27]; however, despite better prevention strategies, cervical cancer is still not sufficiently manageable worldwide with a stagnating mortality rate. Most cancers in the developed world present in early FIGO stage IA1–IIA, while primary metastatic disease is uncommon. Surgery including radical hysterectomy including pelvic lymph node resection for staging is the gold standard. Patients with high-risk features including insufficient margins, large tumors, and lymph vascular space invasion receive adjuvant radiochemotherapy (RCTX) with platinum [28]. From stage IIB onwards, patients are treated with combined radiochemotherapy with platinum. This was established in 1999 when five randomized controlled trials demonstrated a 30–50% survival benefit for patients treated with combined radiochemotherapy compared with radiation alone.

A large meta-analysis of chemoradiation trials demonstrated an absolute overall survival (OS) benefit of 12% [29]. Since these trials, no practice changing studies were published until 2014, when the findings of a phase III study with bevacizumab resulted in its approval for late-stage cervical cancer by the Food and Drug Administration (FDA) and European Medical Agency (EMA). In a large randomized phase III trial, two chemotherapy regimens with cisplatin and paclitaxel or topotecan and paclitaxel plus or minus bevacizumab were examined [30]. Bevacizumab was applied during chemotherapy and as maintenance therapy until disease progression, achieving an increased OS benefit (17.0 months versus 13.3 months; hazard ratio (HR) for death, 0.71; 98% confidence interval (CI), 0.54–0.95; P = 0.004) and a higher response rate (48 versus 36%, P = 0.008). An additional quality of life (QoL) analysis confirmed the low toxicity profile and good tolerability without any deterioration of quality of life [31].

2.1. Immune system and cervical cancer

Most cervical cancers are associated with HPV infection. The cervical epithelium is the ideal area for HPV because of the absence of an inflammatory milieu, which provides a protective niche where the HPV is capable of evading the host immune response for many months. Research has provided some insight into the means of evasion by the virus in cervical cancer [32]. The presence of CD4+ lymphocytes in precursor lesions and CD8+ lymphocytes in malignant tumors in the absence of an effective immune response suggests that T cell cytotoxic responses are impaired [33, 34]. Indeed, the zeta chain of the T-cell receptor is downregulated in CD8+ lymphocytes in cervical cancer, suggesting defective T cell signaling [35]. Furthermore, NKG2D-expressing natural killer and cytotoxic T cells, which have a key role in the elimination of virus-infected and tumor cells, are present at reduced levels in both patients with cervical cancer and cervical intraepithelial neoplasia [36]. Increased T regulatory cell activity has also been reported [37]. The immunoregulatory enzyme, IDO, appears to facilitate the induction of immune escape together with T regulatory cells [38]. Understanding the different mechanisms of immune evasion in cervical cancer is key to establishing new treatments.

2.2. Checkpoint-inhibitors in cervical cancers

Despite this new regime, the prognosis for metastatic and locally advanced cervical cancer is still poor, with an OS of 12–17 months [30, 39]. To improve prognosis, new treatment options are urgently needed. One important strategy is to enable the immune system to reject the tumors facilitating checkpoint-inhibitors. An important strategy to improve T cell-dependent tumor attack is by inhibiting immune T cell checkpoints. A checkpoint-inhibitor is a drug that inhibits certain surface proteins made by specific immune cells, such as T cells and cancer cells. These specific proteins control the immune responses and prevent T cells from killing cancer cells. Inhibiting these proteins will remove the natural surveillance of the immune system and T cells will be activated to eliminate cancer cells.

Blocking inhibiting checkpoints like cytotoxic T-lymphocyte-associated antigen 4 (CTLA-4) or PD-1/PD-L1 results in activation of T cell proliferation and cytokine production. CTLA-4 begins to be expressed on the naïve T cell 48 hours after activation in lymph nodes and is closely associated with attenuation of these activating T cells [40]. PD-1 is expressed on effector T cells in peripheral tissues and binds with PD-L1 (B7-H1) and PD-L2 (B7-DC) expressed on DCs or tumor cells for attenuation of activated effector T cells [41, 42]. Under normal circumstances, interferon (IFN)-γ upregulates the expression of PD-L1, protecting DCs from T cell-mediated cytotoxicity [43]. However, in head and neck squamous cell carcinomas associated with HPV, the number of CD8+ T cells expressing PD-1 has been reported to be higher in tumors than peripheral blood. This suggests that the expression of PD-1 by CD8+ T cells starts after entering into the tumor microenvironment.

Currently, there are several studies in the U.S. and EU examining different checkpoint-inhibitors and combinations, e.g., chemotherapy, PARP-inhibitors, or antiangiogenetic agents, in cervical cancer and other solid tumors (Table 1). Presently, most studied checkpoint-inhibitors are pembrolizumab, nivolumab, ipilimuab, and durvalumab.

ClinicalTrials.gov Identifier:PhaseDrugSituation
NCT02471846IGDC-0919
(small molecule investigational immunotherapy designed to inhibit IDO (Indoleamine 2,3-dioxygenase), a protein often overproduced by many cancer cells)
plus atezolizumab
Metastatic tumors including CC
NCT02812875IBCA-170
Oral small molecule inhibiting PD-L-1/2
Metastatic solid tumors including CC
NCT02635360IIPembrolizumabCombing with RCTX for advanced CC
NCT02834013IINivolumab plus ipilimumabMetastatic rare tumors including CC

Table 1.

Data on immune therapy agents in particular checkpoint-inhibitors in cervical cancer.

Notes: Ongoing checkpoint-inhibitor studies in cervical cancers. RCTX = radiochemotherapy; CC = cervical cancer.

Pembrolizumab, a PD-1 antibody, is approved for patients with advanced melanomas [44]. In cervical cancer, it is currently being investigated in a phase II trial (NCT02628067) based on the phase IB data presented at ASCO 2016 [45]. Patients with stage IVB or nonresectable cervical cancer received 10 mg/kg pembrolizumab every 2 weeks until disease progression or toxicity for a total treatment of 24 months. The overall response rate (ORR) was 17% (95% CI 5–36). While no grade 4 toxicity occurred, two treatment-related discontinuations were observed (grade 3 colitis and grade 3 Guillian-Barré syndrome). The median progression-free survival (PFS) and OS were 2 and 9 months, respectively. The 12 months PFS and OS were 8 and 33%, respectively. Some patients had very long remission rates that are promising and will lead to further evaluation in cervical cancer.

Nivolumab has been approved for metastatic and unresectable lung cancer [46], where it showed a survival benefit compared to conventional chemotherapy treatment. This PD-1 antibody has also been tested in a phase I/II study for patients with HPV-associated tumors, including cervical, vaginal, and vulvar cancer (www.clinicaltrials.gov:NCT02488759).

Ipilimumab is an anti-CTLA-4 antibody and was the first checkpoint inhibitor approved for metastatic melanoma and has significantly improved the OS of this disease [19]. It is at present tested in several other tumors including gynecologic cancers. In cervical cancer, it has been tested in a phase I study following standard radiochemotherapy in patients with locally advanced cervical cancer (www.clinicaltrials.gov: NCT01711515). The trial is currently recruiting patients.

Another phase 2 trial from Princess Margaret Hospital examines the role of ipilimumab in patients with metastatic or recurrent human papillomavirus-related cervical cancer (www.clinicaltrials.gov: NCT01693783).

Durvalumab (MEDI4736), an anti-PD-L1 antibody, is being tested in combination with tremelimumab, an anti-CTLA-4 antibody [47, 48] in a phase I trial for patients with six different types of cancer, including cervical cancer (www.clinicaltrials.gov: NCT01975831). Durvalumab inhibits PD-L1 interaction with PD-1 (IC50 0.1 nM) and CD80/B7.1 (IC50 0.04 nM), allowing T-cells to recognize and kill tumor cells.

Early single agent phase I evaluation in several tumor types, including triple negative breast cancer, showed a disease control rate of 33% and an overall response rate of 10% [49]. There were early (5 weeks) and also durable responses (56+ weeks). PD-L1 expression appears to enrich the response to durvalumab monotherapy. Drug-related events were observed in 46% of patients with 7% of patients reporting a grade ≥3 AE that led in 1% to discontinuation of the treatment. The most common drug-related AEs were fatigue, rash/pruritus, diarrhea, and vomiting.

Advertisement

3. Ovarian cancer

Epithelial ovarian cancer (EOC) is the fifth most common cancer in women and one of the main causes of death in relation to gynecologic cancer worldwide [50]. Ovarian cancer has a poor prognosis, probably as three in four cancers are diagnosed in advanced FIGO stages [51]. The 5-year survival rate is poor, estimated at 20–30% for stage I–IV disease. Not only the tumor stage, but also the histopathological subtype is prognosis defining, with poor differentiated serous cancers having the poorest outcomes [2]. Best prognosis is seen in mucinous subtype. These subtype is mostly localized FIGO stage I disease [52].

Surgery with optimal debulking still has a major influence on the outcome in advanced EOC. Best outcome has been reported for patients achieving maximal cytoreductive surgery without macroscopic residual disease [53].

The most promising novel agents for ovarian cancer are antiangiogenesis-based therapies, e.g., bevacizumab, pazopanib, cediranib, or trebananib and PARP-inhibitors, e.g., olaparib or niraparib [5460]. Bevacizumab and olaparib are approved in the United States and Europe and demonstrated a PFS benefit of 3–4 months when used during or after platinum-based chemotherapy. In BRCA positive patients, there was a PFS benefit of more than 9 months for patients diagnosed with relapsed serous high grade EOC [61].

Recent data suggest that also patients without a BRCA mutation might benefit from a treatment with the PARP-inhibitor niraparib. In this recent study published by Mirza et al., niraparib was also beneficial in non-BRCA mutated patients [62]. They used homologous recombination deficiency (HRD) score to predict response on niraparib. In this study, non-BRCA mutated patients had also significant difference 9.3 versus 3.9 months in PFS.

Non-BRCA mutated and HRD-positive 12.9 versus 3.8 months (HR 0.38).

HRD positive and BRCA wildtype: 9.3 versus 3.7 months, somatic BRCA mutated 20.9 versus 11.0 months, HRD negative 6.9 versus 3.8 (HR 0.58).

3.1. Vaccination strategies for ovarian cancer

A number of methods have been used to enhance immune response in ovarian cancer to improve prognosis, yet, none of these methods have been approved. Several types of vaccination strategies have been tested, e.g.:

  1. Anti-idiotype AB-based vaccination, for example, Abagovomab.

  2. Peptide-/protein-based vaccination, for example, NY-ESO peptides.

  3. Lymphocytes-based vaccination, for example, Autologous LAK plus IL2.

  4. Carbohydrate-based vaccination, for example, MUC1-Sialyl-TN.

  5. DNA plasmid-based vaccination, for example, Poxviral Vector PANVAC-V.

  6. Combination-based vaccination, for example, with sunitinib.

  7. Vaccination-based on dendritic cells, for example, autologous DC pulsed with MUC1-derived peptides or HER-2/neu.

The following paragraph will focus on anti-idiotype AB-based vaccination only as this type is best developed and also phase III data are available.

3.2. Vaccination with idiopathic antibodies: abagovomab and oregovomab

In his theory of clonal selection published in 1974, Neils Jerne described how antibodies (Ab1) generated in response to a particular antigen may themselves be immunogenic [63, 64]. The immunogenic determinants of Ab1 antibodies are termed ‘idiotopes’. Ab1 idiotopes can act as antigens, leading to the development of anti-idiotypic antibodies (Ab2) [64]. As idiotopes are largely located in the highly variable region of the antibody that serves as the antigen-binding site, in some cases Ab2 anti-idiotypic antibodies can mimic antigen structure. Indeed, research has shown that exposure to Ab2 anti-idiotypic antibodies can sometimes induce a more pronounced response than exposure to the antigen itself. Exposure to Ab2 anti-idiotypic antibodies may subsequently stimulate the generation of Ab3 antibodies, some of which target Ab2 idiotopes, and are also capable of binding to the antigen.

Abagovomab (ACA-126) is a murine IgG1k monoclonal antibody (Ab2) with an idiotope that imitates CA125. It is under investigation as an anti-idiotypic vaccine for ovarian cancer. CA125 is a mucin-like transmembrane glycoprotein that is upregulated in ovarian cancer and currently represents the most widely used ovarian cancer biomarker [65, 66]. The biological function of CA125 remains poorly understood, with putative roles in cell adhesion, migration, invasion, and possible immunosuppressive properties suggested [67, 68].

In phase I studies in patients with chemotherapy-resistant ovarian cancer, abagovomab was associated with induction of Ab3 and HAMA responses, increased serum levels of interferon (IFN)-γ, and increases in CA125-specific CD8+ T cells postvaccination [69], suggesting the induction of Th1 immune responses [70]. The induction of Ab3 response was confirmed in a phase Ib/II clinical trial with abagovomab in 119 patients with CA125-positive ovarian, tubal, and peritoneal cancer [71]. Ab3 response occurred in 68.1% of patients and was associated with prolonged overall survival (OS) compared with nonresponders (23.4 versus 4 months; P < 0.0001), regardless of FIGO stage, first-line chemotherapy, or previous treatment. Antibody-dependent cellular cytotoxicity (ADCC), observed in 26.9% of patients, was also associated with significantly prolonged survival (25 versus 10 months; P = 0.0126), suggesting a role for ADCC in the antitumor effect of abagovomab. Nevertheless, abagovomab was not associated with prolonged recurrence-free survival (RFS) or OS compared with placebo when administered as a maintenance therapy to patients (n = 888) with first remission of ovarian cancer (FIGO stage III/IV) during the phase III ‘Monoclonal antibody Immunotherapy for Malignancies of Ovary by Subcutaneous Abagovomab’ (MIMOSA) trial, despite the induction of measurable immune response [72].

Oregovomab (B43.13, OvRex) also targets CA125, binding with high affinity (KD = 1.2 × 1010 M−1). This murine IgG1 monoclonal antibody was investigated for the treatment of ovarian cancer after a survival advantage was noted during its initial use as a technetium 99c-labeled agent for the immunoscintigraphic detection of recurrent ovarian cancer [73].

Infusion of the antibody results in the formation of immune complexes with circulating antigen that trigger generation of anti-CA125 antibodies [74]. Indeed, oregovomab appears to induce broad humoral and cellular anti-CA125 responses.

During a phase I trial, multiple infusions of oregovomab were associated with a greater than threefold increase in anti-CA125 antibody levels in nearly half (43%) of patients (n = 184) with ovarian cancer (FIGO stages I–IV) [74]. Anti-CA125 antibody response was associated with prolonged survival compared with nonresponse (22.9 versus 13.5 months; P = 0.0089), and an increase in T-cell proliferation was noted, which was also associated with prolonged survival. In a phase II study (n = 20), T-cell responses to CA125 and/or autologous tumors were also shown to correlate with prolonged survival in oregovomab-treated patients with platinum-resistant recurrent ovarian cancer (FIGO stages I–IV) compared with nonresponders (median not reached versus 51.9 weeks) [75].

While oregovomab elicits tumor-specific T-cell responses, it does not appear to be able to directly inhibit tumor growth. Anti-CA125 antibodies isolated from oregovomab-treated patients with ovarian cancer (FIGO stages I–IV) in one study were able to mediate ADCC in the presence of peripheral blood mononuclear cells, but not CDC [76].

In addition, there are conflicting data on the association between immune response to oregovomab and clinical benefit. A retrospective analysis of 44 patients with recurrent ovarian cancer (majority FIGO stages III and IV) who received technetium 99c-labeled oregovomab reported a significant relationship between immune response and survival [73]. More than 67% of patients had HAMA and Ab2 responses, with 28% of patients experiencing a more than threefold increase in anti-CA125 antibody levels. These immune responses were associated with prolonged survival compared with nonresponders: HAMA (22.6 versus 7.2 months; P = 0.0016), Ab2 (18.3 versus 9.3 months; P = 0.075), and anti-CA125 (18.2 versus 13.1 months; P = 0.0896). By contrast, no reduction in tumor burden was detected in 13 oregovomab-treated patients with ovarian cancer during a pilot phase II study, despite measurable T- and B-cell responses in the majority of patients [77]. Furthermore, oregovomab monoimmunotherapy was associated with similar clinical outcomes to placebo during a phase III trial in 375 patients with advanced ovarian cancer (FIGO stage III/IV), despite measurable bioactivity [78].

The potential for combining oregovomab with front-line chemotherapy has been investigated during a phase II clinical trial in 40 patients with advanced ovarian cancer (FIGO stages III/IV) [79]. Patients were randomized to receive oregovomab via two dosing schedules: either on the same day as or 1 week after standard carboplatin-paclitaxel chemotherapy. Primary and secondary endpoints compared antibody and cellular response between the two dosing schedules, but the authors also noted that the immune responses triggered were stronger than those observed in previous studies using oregovomab monoimmunotherapy.

3.3. Immune checkpoint-inhibitors in ovarian cancer

Apart from the vaccination, immune checkpoint-inhibitors such as the programmed cell death 1 protein (anti-PD1)/PD-Ligand1 and CTL-A4 were also under research for ovarian cancer [42] (see also Table 2). Tumors with high mutational loads are ideal candidates for therapies with immune checkpoint-inhibitors. In melanoma and lung cancer, these new drugs are already approved [40, 46]. The role of immune checkpoint-inhibitors in ovarian cancer is not so clear so far, although PD1/PD-L1 pathway seems to play an important role in ovarian cancer. In ovarian cancers, PD-1 is expressed on TILs [80]. Expression of PD-L1 on tumors has been shown to be bad prognostic factor [81]. In a preclinical model inhibition of PD1 and PD-L1 demonstrated tumor rejection and reprogramming of tumor microenvironment [82]. In one of first phase I study for an anti-PD-L1 antibody, there were also responses seen for ovarian cancers [83].

DrugTargetPatientsNPD-L1 StatusORR (%)DCR (%)CRPRSDLiterature
NivolumabPD-1Rezidiv platin-resistant18All1744215[84]
PembrolizumabPD-1Fortgeschrittenes EOC26PD-L1+11,534,6126[85]
AvelumabPD-L1Platin/Chemo-resistentes EOC75All10,754,70833[92]
BMS-936559PD-L1Fortgeschrittenes EOC17All123.5013[83]

Table 2.

Data available on immune checkpoint-inhibitors used in ovarian cancer.

Notes: The clinical data for checkpoint-inhibitors mainly anti-PD-1 in EOC. ORR = overall response rate; DCR = disease control rate; PR = partial remission; CR = complete remission; SD = stable disease; EOC = epithelial ovarian cancer.

First, data from phase a phase II studies demonstrated low response rates but a higher disease control rated and long-term remissions. The patients treated in these trials had poor prognostic disease and were platinum resistant [84]. The assessment of PD-L1 as a prognostic marker for ovarian cancer is less clear. The data from Hamannishi et al. demonstrated that PD-1 was not ideal as prognostic marker [81].

More importantly, the mutational landscape might be important to select the right treatment for the suitable tumor. In general, the mutational burden is lower in ovarian cancer than in other cancers. But ovarian tumors with germline or somatic BRCA1/2 mutations were found to have a higher frequency of exome mutations (67.5 on average) than tumors with wild-type BRCA (49.5 on average) [86].

3.3.1. Ipilimumab

Ipilimumab (MDX-CTLA-4, Yervoy) is a full human IgG1 monoclonal antibody to CTLA-4 approved by the FDA for the treatment of advanced melanoma on the basis of phase III observations of prolonged OS (median 4 months versus tumor vaccine) in patients with unresectable pretreated stages III and IV melanoma [40]. Immune response appears to underlie the antitumor effect of ipilimumab. Studies with ipilimumab in melanoma have shown increased absolute lymphocyte counts [87], upregulation of inducible costimulator (ICOS) on CD4+ T cells [88], and enhanced antibody and T-cell responses to cancer-testis antigen NY-ESO-1 that largely correlate with clinical benefit and prolonged survival [88, 89].

Ipilimumab has also been investigated in a small number of patients with ovarian cancer. Findings from two studies in a total of 11 patients with previously vaccinated ovarian cancer (FIGO stage IV) suggest that ipilimumab is generally well tolerated and can trigger a decrease/stabilization of CA125 [90, 91]. Significant antitumor effects were observed in some patients. One patient experienced a marked reduction in serum CA125 levels during ipilimumab treatment with a substantial regression of a large cystic hepatic metastasis, complete resolution of mesenteric lymphadenopathy, and gastrocolic ligament thickening [91]. Increased antibody responses to NY-ESO-1, which is expressed in many ovarian carcinomas, were also detectable and correlated with therapeutic activity. Four additional patients achieved stable disease.

3.3.2. Avelumab

Avelumab (MSB0010718C; anti-PD-L1) is a full human anti-PD-L1 IgG1 antibody currently under clinical investigation for several cancers. It was tested in a phase IB study for chemotherapy refractory EOC. Safety and clinical activity data were reported at ASCO 2016 [92]. Patients with advanced EOC unselected for PD-L1 expression received avelumab 10 mg/kg IV every 2 weeks until progression, unacceptable toxicity, or withdrawal. Tumors were assessed every 6 weeks according to RECIST 1.1. Unconfirmed ORR, PFS, and OS were evaluated and AE graded by NCI-CTCAE v4.0. During a median of 12 weeks (range: 2–54 weeks), 124 patients were treated with AE only occurring in 82 patients (66.1%); most common ones (≥10%) were fatigue (13.7%), infusion-related reaction (12.1%), and diarrhea (11.3%). The ORR was 9.7%. The rate of stable disease was 44.4%. The disease control rate was 54.0%. PD-L1 expression was evaluable in 74 patients with PD-L1+ tumors expressing an ORR of 12.3% and in PD-L1 tumors of 5.9%.

In first line and maintenance setting, avelumab is currently evaluated in a phase III, open-label, international, multicenter study as additional maintenance therapy after debulking surgery (www.clinicaltrials.gov: NCT02718417). The study has three arms. Arm A includes chemotherapy with carboplatin plus paclitaxel (standard of care), arm B includes chemotherapy followed by avelumab in maintenance, and arm C includes chemotherapy in combination with avelumab followed by avelumab in maintenance. The primary endpoint of this study is PFS.

In this platinum-resistant and refractory setting, avelumab is combined with PEGylated doxorubicin versus single-agent PEGylated doxorubicin or avelumab alone (www.clinicaltrials.gov: NCT02580058). The primary endpoint of this study is OS.

Advertisement

4. Vulvar cancers

With approximately 4% of the tumors of the female genital tract, vulvar carcinoma is rare. In 75% of cases, it occurs as type 2 carcinoma in the elderly patient. The median age is 70 years. As type 1 carcinoma, which is usually associated with HPV, it occurs in younger patients in combination with cervical carcinoma or anal carcinoma. The most important treatment is surgical resection. In approximately 30% of the cases, a complete R0 resection is not possible, and then combined procedures are used before or after surgery with chemotherapy and radiotherapy. For systemic approaches, in general, platinum-based chemotherapy regimens are used, frequently in combination with 5FU, mitomycine-c, taxanes, and ifosfamide [93].

Prognosis for vulvar cancer is still poor and there is a high recurrence rate. The 10-year survival rate is still modest with 65% [1]. Therefore, new treatment options are urgently needed. Standard of care for metastatic situations remains the standard platinum-based chemotherapy [94]. Newer targeted therapy failed to demonstrate a major survival benefit [95]. Immunotherapy especially with checkpoint-inhibitors might therefore be beneficial for squamous cell cancers of the female genital tract.

In anal cancer early data from the first 37 patients having received nivolumab every 2 weeks have recently been presented [96]. Here, two patients (5%) showed a complete response, seven (19%) had a partial response, and 17 (46%) had stable disease. The disease control rate was high with 79% and a median PFS of 3.9 months with 6 patients still remaining on the study at present. However, side effects included fatigue, anemia, rash, and one incident of pneumonitis. For vulvar cancers, currently three trials incorporate checkpoint inhibitors worldwide (www.clinicaltrials.gov: NCT 02858310, NCT02628067, NCT02834013).

Advertisement

5. Endometrial cancers

In the group of gynecologic cancers, endometrial cancers have the best outcome with a five-year overall survival rate of 80% [97]. In general, the disease can be cured with surgery including staging procedures and radiotherapy [98, 99]. For type II cancer and advanced stage disease, chemotherapy with carboplatin and paclitaxel is included in standard of care treatment [98, 99]. For relapsed disease and stage IV disease, surgery can be applied, but in general, standard of care is systemic therapy, including endocrine therapy such as medroxy-progestine, tamoxifen, or aromatase inhibitors [100]. Furthermore, combination chemotherapy with carboplatin and paclitaxel is used [101]. For further progression, agents like doxorubicin or topotecan have minimal activity [102, 103].

For advanced and relapsed endometrial cancer, new treatment options are urgently needed. Beside targeted therapy including combinations with endocrine therapy and CDK4/6 inhibitors or VEGF-(R) inhibitors or FGFR-inhibitors, application of immunotherapy might have strong impact in that stage of disease [104, 105].

The cancer genome atlas has recently classified endometrial cancers in four distinct subgroups [106, 107]:

  1. POLE-ultramutated.

  2. Microsatellite instability (MSI) hypermutated.

  3. Copy number low.

  4. Copy number high.

In endometrial cancers, there are only few data available for a specific immunotherapeutic approach, despite the knowledge that high mutational load tumors are expected to respond well. There are some data about dendritic cell vaccination [108110] and about checkpoint inhibition, particularly in view of mismatch repair-deficient cancers [111]. In this phase II study, the authors examined the efficacy of pembrolizumab in several tumor types (n = 41), including two endometrial cancers. For mismatch repair-deficient colorectal cancers, the immune-related objective response rate was 40 versus 0% for mismatch-repair-proficient colorectal cancers. Patients with other mismatch repair-deficient tumors had comparable response rates. Whole-exome sequencing showed a much higher rate of somatic mutations for mismatch repair-deficient tumors (mean number of somatic mutations: 1782 versus 73 in mismatch repair-proficient tumors (P = 0.007)). The noncolorectal cohort included nine mismatch repair-deficient cancers including gastric cancer, ampullar or cholangiocarcinoma, small bowel cancer, and endometrial cancer. The objective response rate was 71% (95% CI 29—96) with a median time to response of 12 weeks (95% CI 10–13 months). The treatment was well tolerated; most common side effects (all grades) were rash, pruritus, diarrhoea, allergic rhinitis, and pain. The authors conclude that the treatment was well tolerated and the evaluation of mismatch repair deficiency might be a useful marker, independent of underlying tumor type.

Advertisement

6. Conclusion

There has been a tremendous success for immunotherapy in certain tumor types (e.g., melanoma, lung cancer etc.), in particular, for immune checkpoint-inhibitors. In gynecological cancers, the situation is less clear although there are some promising data, especially for treatment with anti-PD1 or anti-PD-L1 antibodies. Early clinical trials showed encouraging disease control rates in heavily pretreated patients. A combination of checkpoint-inhibitors, e.g., anti-PD-1 with CTLA-4 or anti-PD-1 with LAG3 or combinations with chemotherapy might overcome resistance in this type of disease. Current trials aim to examine the combination between immune checkpoint-inhibitors and VEGF inhibitors like bevacizumab or PARP-inhibitors like olaparib and niraparib. An important role of these combination trials is to improve quality of life for patients. Another important goal is the incorporation of appropriate biomarkers to identify new immunotherapeutic approaches. The situation about immunotherapy in other than ovarian cancer has to be called scarce, and no conclusion can be drawn from the data in these cancers up to date.

Advertisement

Abbreviations

AE

Adverse event

AB

Antibody

CAR

T-cell receptor or chimeric antigen receptor

CC

Cervical cancer

CD

Cluster of differentiation

CR

Complete remission

DC

Dendritic cells

EMA

European Medical Agency

EOC

Epithelial ovarian cancer

FDA

US Food and Drug Administration

FIGO

International Federation of Gynecology and Obstetrics

HPV

Human papillomavirus

IC50

Half maximal inhibitory concentration

IFN-α

Interferon-alpha

IFN-γ

Interferon-gamma

nM

Nanomol

ORR

Overall response rate

OS

Overall survival

PD-1

Programmed death cell ligand 1

PFS

Progression free survival

PR

Partial remission

QoL

Quality of life

RCTX

Radiochemotherapy

RR

Response rate

TLR

Toll-like receptor

References

  1. 1. American Cancer Society. Cancer Facts & Figures 2015. Atlanta: American Cancer Society; 2015. https://old.cancer.org/acs/groups/content/@editorial/documents/document/acspc-044552.pdf
  2. 2. Winter WE, Maxwell GL, Tian C, Carlson JW, Ozols RF, Rose PG, et al. Prognostic factors for stage III epithelial ovarian cancer: a Gynecologic Oncology Group Study. J Clin Oncol [Internet]. 2007;25(24):3621–7. Available from: http://www.ncbi.nlm.nih.gov/pubmed/17704411
  3. 3. Bourla AB, Zamarin D. Immunotherapy: new strategies for the treatment of gynecologic malignancies. Oncology (Williston Park) [Internet]. 2016 Jan;30(1):59–66, 69. Available from: http://www.ncbi.nlm.nih.gov/pubmed/26791846
  4. 4. Pfisterer J, Harter P, Simonelli C, Peters M, Berek J, Sabbatini P, et al. Abagovomab for ovarian cancer. Expert Opin Biol Ther. 2011;11:395–403.
  5. 5. Zhang L, Conejo-Garcia JR, Katsaros D, Gimotty PA, Massobrio M, Regnani G, et al. Intratumoral T cells, recurrence, and survival in epithelial ovarian cancer. N Engl J Med. 2003;348:203–13.
  6. 6. Hwang W-T, Adams SF, Tahirovic E, Hagemann IS, Coukos G. Prognostic significance of tumor-infiltrating T cells in ovarian cancer: a meta-analysis. Gynecol Oncol [Internet]. 2012 Feb;124(2):192–8. Available from: http://www.ncbi.nlm.nih.gov/pubmed/22040834
  7. 7. Sato E, Olson SH, Ahn J, Bundy B, Nishikawa H, Qian F, et al. Intraepithelial CD8+ tumor-infiltrating lymphocytes and a high CD8+/regulatory T cell ratio are associated with favorable prognosis in ovarian cancer. Proc Natl Acad Sci U S A. 2005;102:18538–43.
  8. 8. Smyth MJ, Dunn GP, Schreiber RD. Cancer immunosurveillance and immunoediting: the roles of immunity in suppressing tumor development and shaping tumor immunogenicity. Adv Immunol [Internet]. 2006;90:1–50. Available from: http://www.ncbi.nlm.nih.gov/pubmed/16730260
  9. 9. Herold M, Dölken G, Fiedler F, Franke A, Freund M, Helbig W, et al. Randomized phase III study for the treatment of advanced indolent non-Hodgkin’s lymphomas (NHL) and mantle cell lymphoma: chemotherapy versus chemotherapy plus rituximab. Ann Hematol [Internet]. 2003 Feb;82(2):77–9. Available from: http://www.ncbi.nlm.nih.gov/pubmed/12601483
  10. 10. Chen DS, Mellman I. Oncology meets immunology: the cancer-immunity cycle. Immunity [Internet]. 2013 Jul 25;39(1):1–10. Available from: http://www.ncbi.nlm.nih.gov/pubmed/23890059
  11. 11. Leffers N, Daemen T, Helfrich W, Boezen HM, Cohlen BJ, Melief CJM, et al. Antigen-specific active immunotherapy for ovarian cancer. Cochrane Database Syst Rev [Internet]. 2014 Sep 17;(9):CD007287. Available from: http://www.ncbi.nlm.nih.gov/pubmed/25229990
  12. 12. Edwards RP, Gooding W, Lembersky BC, Colonello K, Hammond R, Paradise C, et al. Comparison of toxicity and survival following intraperitoneal recombinant interleukin-2 for persistent ovarian cancer after platinum: twenty-four-hour versus 7-day infusion. J Clin Oncol [Internet]. 1997 Nov;15(11):3399–407. Available from: http://www.ncbi.nlm.nih.gov/pubmed/9363872
  13. 13. 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. Gynecol Oncol [Internet]. 2014 Jun;133(3):433–8. Available from: http://www.ncbi.nlm.nih.gov/pubmed/24708919
  14. 14. Rosenberg SA, Restifo NP. Adoptive cell transfer as personalized immunotherapy for human cancer. Science [Internet]. 2015 Apr 3;348(6230):62–8. Available from: http://www.ncbi.nlm.nih.gov/pubmed/25838374
  15. 15. Lee DW, Kochenderfer JN, Stetler-Stevenson M, Cui YK, Delbrook C, Feldman SA, et al. T cells expressing CD19 chimeric antigen receptors for acute lymphoblastic leukaemia in children and young adults: a phase 1 dose-escalation trial. Lancet (London, England) [Internet]. 2015 Feb 7;385(9967):517–28. Available from: http://www.ncbi.nlm.nih.gov/pubmed/25319501
  16. 16. Robbins PF, Kassim SH, Tran TLN, Crystal JS, Morgan RA, Feldman SA, et al. A pilot trial using lymphocytes genetically engineered with an NY-ESO-1-reactive T-cell receptor: long-term follow-up and correlates with response. Clin Cancer Res [Internet]. 2015 Mar 1;21(5):1019–27. Available from: http://www.ncbi.nlm.nih.gov/pubmed/25538264
  17. 17. Curiel TJ, Coukos G, Zou L, Alvarez X, Cheng P, Mottram P, et al. Specific recruitment of regulatory T cells in ovarian carcinoma fosters immune privilege and predicts reduced survival. Nat Med [Internet]. 2004 Sep;10(9):942–9. Available from: http://www.ncbi.nlm.nih.gov/pubmed/15322536
  18. 18. 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. Int J cancer [Internet]. 2014 Jan 1;134(1):32–42. Available from: http://www.ncbi.nlm.nih.gov/pubmed/23784932
  19. 19. Chevolet I, Speeckaert R, Schreuer M, Neyns B, Krysko O, Bachert C, et al. Characterization of the in vivo immune network of IDO, tryptophan metabolism, PD-L1, and CTLA-4 in circulating immune cells in melanoma. Oncoimmunology [Internet]. 2015 Mar;4(3):e982382. Available from: http://www.ncbi.nlm.nih.gov/pubmed/25949897
  20. 20. Wainwright DA, Chang AL, Dey M, Balyasnikova I V, Kim CK, Tobias A, et al. Durable therapeutic efficacy utilizing combinatorial blockade against IDO, CTLA-4, and PD-L1 in mice with brain tumors. Clin Cancer Res [Internet]. 2014 Oct 15;20(20):5290–301. Available from: http://www.ncbi.nlm.nih.gov/pubmed/24691018
  21. 21. Siegel R, Ma J, Zou Z, Jemal A. Cancer statistics, 2014. CA Cancer J Clin. 2014;64(1):9–29.
  22. 22. Schiffman M, Solomon D. Clinical practice. Cervical-cancer screening with human papillomavirus and cytologic cotesting. N Engl J Med [Internet]. 2013 Dec 12;369(24):2324–31. Available from: http://www.ncbi.nlm.nih.gov/pubmed/24328466
  23. 23. Muñoz N, Bosch FX, de Sanjosé S, Herrero R, Castellsagué X, Shah KV, et al. Epidemiologic classification of human papillomavirus types associated with cervical cancer. N Engl J Med [Internet]. 2003 Feb 6;348(6):518–27. Available from: http://www.ncbi.nlm.nih.gov/pubmed/12571259
  24. 24. Choi YJ, Park JS. Clinical significance of human papillomavirus genotyping. J Gynecol Oncol [Internet]. 2016 Mar;27(2):e21. Available from: http://www.ncbi.nlm.nih.gov/pubmed/26768784
  25. 25. Krebsliga Schweiz, NICER Report. Krebs in der Schweiz: wichtige Zahlen. 2012;2008: 2005–9. http://www.nicer.org/en/publications/scientific-publications/#content1957
  26. 26. Kahn JA. HPV vaccination for the prevention of cervical intraepithelial neoplasia. N Engl J Med [Internet]. 2009 Jul 16;361(3):271–8. Available from: http://www.ncbi.nlm.nih.gov/pubmed/19605832
  27. 27. Vesikari T, Brodszki N, van Damme P, Diez-Domingo J, Icardi G, Petersen LK, et al. A randomized, double-blind, phase III study of the immunogenicity and safety of a 9-valent human papillomavirus L1 virus-like particle vaccine (V503) versus Gardasil® in 9-15-year-old girls. Pediatr Infect Dis J [Internet]. 2015 Sep;34(9):992–8. Available from: http://www.ncbi.nlm.nih.gov/pubmed/26090572
  28. 28. Koh W-J, Greer BE, Abu-Rustum NR, Apte SM, Campos SM, Cho KR, et al. Cervical cancer, Version 2.2015. J Natl Compr Canc Netw [Internet]. 2015 Apr;13(4):395–404; quiz 404. Available from: http://www.ncbi.nlm.nih.gov/pubmed/25870376
  29. 29. Green JA, Kirwan JM, Tierney JF, Symonds P, Fresco L, Collingwood M, et al. Survival and recurrence after concomitant chemotherapy and radiotherapy for cancer of the uterine cervix: a systematic review and meta-analysis. Lancet (London, England) [Internet]. 2001 Sep 8;358(9284):781–6. Available from: http://www.ncbi.nlm.nih.gov/pubmed/11564482
  30. 30. Tewari KS, Sill MW, Long HJ, Penson RT, Huang H, Ramondetta LM, et al. Improved survival with bevacizumab in advanced cervical cancer. N Engl J Med [Internet]. 2014 Feb 20;370(8):734–43. Available from: http://www.ncbi.nlm.nih.gov/pubmed/24552320
  31. 31. Penson RT, Huang HQ, Wenzel LB, Monk BJ, Stockman S, Long HJ, et al. Bevacizumab for advanced cervical cancer: patient-reported outcomes of a randomised, phase 3 trial (NRG Oncology-Gynecologic Oncology Group protocol 240). Lancet Oncol [Internet]. 2015 Mar;16(3):301–11. Available from: http://www.ncbi.nlm.nih.gov/pubmed/25638326
  32. 32. Patel S, Chiplunkar S. Host immune responses to cervical cancer. Curr Opin Obstet Gynecol [Internet]. 2009 Feb;21(1):54–9. Available from: http://www.ncbi.nlm.nih.gov/pubmed/19125004
  33. 33. Monnier-Benoit S, Mauny F, Riethmuller D, Guerrini J-S, Căpîlna M, Félix S, et al. Immunohistochemical analysis of CD4+ and CD8+ T-cell subsets in high risk human papillomavirus-associated pre-malignant and malignant lesions of the uterine cervix. Gynecol Oncol [Internet]. 2006 Jul;102(1):22–31. Available from: http://www.ncbi.nlm.nih.gov/pubmed/16427684
  34. 34. de Jong A, van Poelgeest MIE, van der Hulst JM, Drijfhout JW, Fleuren GJ, Melief CJM, et al. Human papillomavirus type 16-positive cervical cancer is associated with impaired CD4+ T-cell immunity against early antigens E2 and E6. Cancer Res [Internet]. 2004 Aug 1;64(15):5449–55. Available from: http://www.ncbi.nlm.nih.gov/pubmed/15289354
  35. 35. Santin AD, Bellone S, Palmieri M, Bossini B, Roman JJ, Cannon MJ, et al. Induction of tumor-specific cytotoxicity in tumor infiltrating lymphocytes by HPV16 and HPV18 E7-pulsed autologous dendritic cells in patients with cancer of the uterine cervix. Gynecol Oncol [Internet]. 2003 May;89(2):271–80. Available from: http://www.ncbi.nlm.nih.gov/pubmed/12713991
  36. 36. Arreygue-Garcia NA, Daneri-Navarro A, del Toro-Arreola A, Cid-Arregui A, Gonzalez-Ramella O, Jave-Suarez LF, et al. Augmented serum level of major histocompatibility complex class I-related chain A (MICA) protein and reduced NKG2D expression on NK and T cells in patients with cervical cancer and precursor lesions. BMC Cancer [Internet]. 2008 Jan 21;8:16. Available from: http://www.ncbi.nlm.nih.gov/pubmed/18208618
  37. 37. Visser J, Nijman HW, Hoogenboom B-N, Jager P, van Baarle D, Schuuring E, et al. Frequencies and role of regulatory T cells in patients with (pre)malignant cervical neoplasia. Clin Exp Immunol [Internet]. 2007 Nov;150(2):199–209. Available from: http://www.ncbi.nlm.nih.gov/pubmed/17937675
  38. 38. Nakamura T, Shima T, Saeki A, Hidaka T, Nakashima A, Takikawa O, et al. Expression of indoleamine 2, 3-dioxygenase and the recruitment of Foxp3-expressing regulatory T cells in the development and progression of uterine cervical cancer. Cancer Sci [Internet]. 2007 Jun;98(6):874–81. Available from: http://www.ncbi.nlm.nih.gov/pubmed/17433037
  39. 39. Monk BJ, Sill MW, McMeekin DS, Cohn DE, Ramondetta LM, Boardman CH, et al. Phase III trial of four cisplatin-containing doublet combinations in stage IVB, recurrent, or persistent cervical carcinoma: a Gynecologic Oncology Group study. J Clin Oncol [Internet]. 2009 Oct 1;27(28):4649–55. Available from: http://www.ncbi.nlm.nih.gov/pubmed/19720909
  40. 40. Hodi FS, O’Day SJ, McDermott DF, Weber RW, Sosman JA, Haanen JB, et al. Improved survival with ipilimumab in patients with metastatic melanoma. N Engl J Med [Internet]. 2010 Aug 19;363(8):711–23. Available from: http://www.ncbi.nlm.nih.gov/pubmed/20525992
  41. 41. Ribas A. Tumor immunotherapy directed at PD-1. N Engl J Med [Internet]. 2012 Jun 28;366(26):2517–9. Available from: http://www.ncbi.nlm.nih.gov/pubmed/22658126
  42. 42. Topalian SL, Hodi FS, Brahmer JR, Gettinger SN, Smith DC, McDermott DF, et al. Safety, activity, and immune correlates of anti-PD-1 antibody in cancer. N Engl J Med [Internet]. 2012 Jun 28;366(26):2443–54. Available from: http://www.ncbi.nlm.nih.gov/pubmed/22658127
  43. 43. 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 Res [Internet]. 2013 Mar 15;73(6):1733–41. Available from: http://www.ncbi.nlm.nih.gov/pubmed/23288508
  44. 44. Ribas A, Puzanov I, Dummer R, Schadendorf D, Hamid O, Robert C, et al. Pembrolizumab versus investigator-choice chemotherapy for ipilimumab-refractory melanoma (KEYNOTE-002): a randomised, controlled, phase 2 trial. Lancet Oncol [Internet]. 2015 Aug;16(8):908–18. Available from: http://www.ncbi.nlm.nih.gov/pubmed/26115796
  45. 45. Frenel J-S, Le Tourneau C, O’Neil BH, Ott PA, Piha-Paul SA, Gomez-Roca CA, Van Brummelen E, Rugo HS, Thomas S, Saraf S, Chen M, Varga A. Institut de Cancerologie de l’ F. Pembrolizumab in patients with advanced cervical squamous cell cancer: preliminary results from the phase Ib KEYNOTE-028 study. J Clin Oncol. 2016;34(suppl; abstr 5515).
  46. 46. Borghaei H, Paz-Ares L, Horn L, Spigel DR, Steins M, Ready NE, et al. Nivolumab versus docetaxel in advanced nonsquamous non-small-cell lung cancer. N Engl J Med [Internet]. 2015 Oct 22;373(17):1627–39. Available from: http://www.ncbi.nlm.nih.gov/pubmed/26412456
  47. 47. Antonia S, Goldberg SB, Balmanoukian A, Chaft JE, Sanborn RE, Gupta A, et al. Safety and antitumour activity of durvalumab plus tremelimumab in non-small cell lung cancer: a multicentre, phase 1b study. Lancet Oncol [Internet]. 2016 Mar;17(3):299–308. Available from: http://www.ncbi.nlm.nih.gov/pubmed/26858122
  48. 48. Planchard D, Yokoi T, McCleod MJ, Fischer JR, Kim Y-C, Ballas M, et al. A phase III study of durvalumab (MEDI4736) with or without tremelimumab for previously treated patients with advanced NSCLC: rationale and protocol design of the ARCTIC Study. Clin Lung Cancer [Internet]. 2016 May;17(3):232–236.e1. Available from: http://www.ncbi.nlm.nih.gov/pubmed/27265743
  49. 49. Segal NH, Hamid O, Hwu W, Massard C, Butler M, Antonia SJ, Blake-Haskins A, Robbins PB, Li X, Vasselli JSK. A phase I multi-arm dose-expansion study of the anti-programmed cell death-ligand-1 (PD-L1) antibody MEDI4736: preliminary data. Ann Oncol 25 iv361–72 101093/annonc/mdu342.
  50. 50. Siegel R, Miller K, Jemal A. Cancer statistics, 2015. CA Cancer J Clin. 2015;65(1):29.
  51. 51. Vergote I, Trope CG, Amant F, Kristensen GB, Ehlen T, Johnson N, et al. Neoadjuvant chemotherapy or primary surgery in stage IIIC or IV ovarian cancer. N Engl J Med. United States; 2010 Sep;363(10):943–53.
  52. 52. Heinzelmann-Schwarz VA, Gardiner-Garden M, Henshall SM, Scurry JP, Scolyer RA, Smith AN, et al. A distinct molecular profile associated with mucinous epithelial ovarian cancer. Br J Cancer [Internet]. 2006 Mar 27;94(6):904–13. Available from: http://www.ncbi.nlm.nih.gov/pubmed/16508639
  53. 53. du Bois A, Reuss A, Pujade-Lauraine E, Harter P, Ray-Coquard I, Pfisterer J. Role of surgical outcome as prognostic factor in advanced epithelial ovarian cancer: a combined exploratory analysis of 3 prospectively randomized phase 3 multicenter trials: by the Arbeitsgemeinschaft Gynaekologische Onkologie Studiengruppe Ovarialkarzin. Cancer. United States; 2009 Mar;115(6):1234–44.
  54. 54. du Bois A, Floquet A, Kim J-W, Rau J, del Campo JM, Friedlander M, et al. Incorporation of pazopanib in maintenance therapy of ovarian cancer. J Clin Oncol [Internet]. 2014;32(30):3374–82. Available from: http://www.ncbi.nlm.nih.gov/pubmed/25225436
  55. 55. Eichbaum M, Mayer C, Eickhoff R, Bischofs E, Gebauer G, Fehm T, et al. The PACOVAR-trial: a phase I/II study of pazopanib (GW786034) and cyclophosphamide in patients with platinum-resistant recurrent, pre-treated ovarian cancer. BMC Cancer [Internet]. 2011;11:453. Available from: http://www.ncbi.nlm.nih.gov/pubmed/22014006
  56. 56. Pignata S, Lorusso D, Scambia G, Sambataro D, Tamberi S, Cinieri S, et al. Pazopanib plus weekly paclitaxel versus weekly paclitaxel alone for platinum-resistant or platinum-refractory advanced ovarian cancer (MITO 11): a randomised, open-label, phase 2 trial. Lancet Oncol [Internet]. 2015;16(5):561–8. Available from: http://www.ncbi.nlm.nih.gov/pubmed/25882986
  57. 57. Burger RA, Brady MF, Bookman MA, Fleming GF, Monk BJ, Huang H, et al. Incorporation of bevacizumab in the primary treatment of ovarian cancer. N Engl J Med [Internet]. 2011;365(26):2473–83. Available from: http://www.ncbi.nlm.nih.gov/pubmed/22204724
  58. 58. Pujade-Lauraine E, Hilpert F, Weber B, Reuss A, Poveda A, Kristensen G, et al. Bevacizumab combined with chemotherapy for platinum-resistant recurrent ovarian cancer: The AURELIA open-label randomized phase III trial. J Clin Oncol [Internet]. 2014;32(13):1302–8. Available from: http://www.ncbi.nlm.nih.gov/pubmed/24637997
  59. 59. Monk BJ, Poveda A, Vergote I, Raspagliesi F, Fujiwara K, Bae D-S, et al. Final results of a phase 3 study of trebananib plus weekly paclitaxel in recurrent ovarian cancer (TRINOVA-1): Long-term survival, impact of ascites, and progression-free survival-2. Gynecol Oncol [Internet]. 2016 Oct;143(1):27–34. Available from: http://www.ncbi.nlm.nih.gov/pubmed/27546885
  60. 60. Ledermann J, Harter P, Gourley C, Friedlander M, Vergote I, Rustin G, et al. Olaparib maintenance therapy in platinum-sensitive relapsed ovarian cancer. N Engl J Med. 2012;366(15):1382–92.
  61. 61. Ledermann J, Harter P, Gourley C, Friedlander M, Vergote I, Rustin G, et al. Olaparib maintenance therapy in patients with platinum-sensitive relapsed serous ovarian cancer: a preplanned retrospective analysis of outcomes by BRCA status in a randomised phase 2 trial. Lancet Oncol [Internet]. 2014;15(8):852–61. Available from: http://www.ncbi.nlm.nih.gov/pubmed/24882434
  62. 62. Mirza MR, Monk BJ, Herrstedt J, Oza AM, Mahner S, Redondo A, et al. Niraparib maintenance therapy in platinum-sensitive, recurrent ovarian cancer. N Engl J Med [Internet]. 2016 Oct 7; Available from: http://www.ncbi.nlm.nih.gov/pubmed/27717299
  63. 63. Jerne NK. Clonal selection in a lymphocyte network. Soc Gen Physiol Ser. 1974;29:29–48.
  64. 64. Tse BWC, Collins A, Oehler MK, Zippelius A, Heinzelmann-Schwarz VA. Antibody-based immunotherapy for ovarian cancer: where are we at? Ann Oncol Off J Eur Soc Med Oncol [Internet]. 2014 Feb;25(2):322–31. Available from: http://www.ncbi.nlm.nih.gov/pubmed/24285017
  65. 65. Rustin GJ, Marples M, Nelstrop AE, Mahmoudi M, Meyer T. Use of CA-125 to define progression of ovarian cancer in patients with persistently elevated levels. J Clin Oncol [Internet]. 2001 Oct 15;19(20):4054–7. Available from: http://www.ncbi.nlm.nih.gov/pubmed/11600607
  66. 66. Bast RC. CA 125 and the detection of recurrent ovarian cancer: a reasonably accurate biomarker for a difficult disease. Cancer [Internet]. 2010 Jun 15;116(12):2850–3. Available from: http://www.ncbi.nlm.nih.gov/pubmed/20564390
  67. 67. Gubbels JAA, Belisle J, Onda M, Rancourt C, Migneault M, Ho M, et al. Mesothelin-MUC16 binding is a high affinity, N-glycan dependent interaction that facilitates peritoneal metastasis of ovarian tumors. Mol Cancer [Internet]. 2006;5(1):50. Available from: http://www.ncbi.nlm.nih.gov/pubmed/17067392
  68. 68. Patankar MS, Jing Y, Morrison JC, Belisle JA, Lattanzio FA, Deng Y, et al. Potent suppression of natural killer cell response mediated by the ovarian tumor marker CA125. Gynecol Oncol [Internet]. 2005 Dec;99(3):704–13. Available from: http://www.ncbi.nlm.nih.gov/pubmed/16126266
  69. 69. Sabbatini P, Dupont J, Aghajanian C, Derosa F, Poynor E, Anderson S, et al. Phase I study of abagovomab in patients with epithelial ovarian, fallopiant ube, or primary peritoneal cancer. In Vitro. 2006;12:5503–10.
  70. 70. Pfisterer J, du Bois a., Sehouli J, Loibl S, Reinartz S, Reuß A, et al. The anti-idiotypic antibody abagovomab in patients with recurrent ovarian cancer. A phase I trial of the AGO-OVAR. Ann Oncol. 2006;17:1568–77.
  71. 71. Reinartz S, Köhler S, Schlebusch H, Krista K, Giffels P, Renke K, et al. Vaccination of patients with advanced ovarian carcinoma with the anti-idiotype ACA125: immunological response and survival (phase Ib/II). Clin Cancer Res [Internet]. 2004 Mar 1;10(5):1580–7. Available from: http://www.ncbi.nlm.nih.gov/pubmed/15014007
  72. 72. Sabbatini P, Harter P, Scambia G, Sehouli J, Meier W, Wimberger P, et al. Abagovomab as maintenance therapy in patients with epithelial ovarian cancer: a phase III trial of the AGO OVAR, COGI, GINECO, and GEICO-the MIMOSA study. J Clin Oncol. 2013;31(12):1554–61.
  73. 73. Möbus 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 [Internet]. 2003 Jul;189(1):28–36. Available from: http://www.ncbi.nlm.nih.gov/pubmed/12861134
  74. 74. 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 Biother Radiopharm [Internet]. 2001 Jun;16(3):187–203. Available from: http://www.ncbi.nlm.nih.gov/pubmed/11471484
  75. 75. Gordon AN, Schultes BC, Gallion H, Edwards R, Whiteside TL, Cermak JM, et al. CA125- and tumor-specific T-cell responses correlate with prolonged survival in oregovomab-treated recurrent ovarian cancer patients. Gynecol Oncol [Internet]. 2004 Aug;94(2):340–51. Available from: http://www.ncbi.nlm.nih.gov/pubmed/15297171
  76. 76. Schultes BC, Baum RP, Niesen A, Noujaim AA, Madiyalakan R. Anti-idiotype induction therapy: anti-CA125 antibodies (Ab3) mediated tumor killing in patients treated with Ovarex mAb B43.13 (Ab1). Cancer Immunol Immunother [Internet]. 1998 Jun;46(4):201–12. Available from: http://www.ncbi.nlm.nih.gov/pubmed/9671143
  77. 77. Ehlen TG, Hoskins PJ, Miller D, Whiteside TL, Nicodemus CF, Schultes BC, et al. A pilot phase 2 study of oregovomab murine monoclonal antibody to CA125 as an immunotherapeutic agent for recurrent ovarian cancer. Int J Gynecol Cancer [Internet]. 15(6):1023–34. Available from: http://www.ncbi.nlm.nih.gov/pubmed/16343178
  78. 78. Berek JS, Taylor PT, Gordon A, Cunningham MJ, Finkler N, Orr J, et al. Randomized, placebo-controlled study of oregovomab for consolidation of clinical remission in patients with advanced ovarian cancer. J Clin Oncol. 2004;22(17):3507–16.
  79. 79. Braly P, Nicodemus CF, Chu C, Collins Y, Edwards R, Gordon A, et al. The Immune adjuvant properties of front-line carboplatin-paclitaxel: a randomized phase 2 study of alternative schedules of intravenous oregovomab chemoimmunotherapy in advanced ovarian cancer. J Immunother [Internet]. 2009 Jan;32(1):54–65. Available from: http://www.ncbi.nlm.nih.gov/pubmed/19307994
  80. 80. Matsuzaki J, Gnjatic S, Mhawech-Fauceglia P, Beck A, Miller A, Tsuji T, et al. Tumor-infiltrating NY-ESO-1-specific CD8+ T cells are negatively regulated by LAG-3 and PD-1 in human ovarian cancer. Proc Natl Acad Sci U S A [Internet]. 2010 Apr 27;107(17):7875–80. Available from: http://www.ncbi.nlm.nih.gov/pubmed/20385810
  81. 81. 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. Proc Natl Acad Sci U S A [Internet]. 2007 Feb 27;104(9):3360–5. Available from: http://www.ncbi.nlm.nih.gov/pubmed/17360651
  82. 82. Duraiswamy J, Freeman GJ, Coukos G. Therapeutic PD-1 pathway blockade augments with other modalities of immunotherapy T-cell function to prevent immune decline in ovarian cancer. Cancer Res [Internet]. 2013 Dec 1;73(23):6900–12. Available from: http://www.ncbi.nlm.nih.gov/pubmed/23975756
  83. 83. Brahmer JR, Tykodi SS, Chow LQM, Hwu W-J, Topalian SL, Hwu P, et al. Safety and activity of anti-PD-L1 antibody in patients with advanced cancer. N Engl J Med [Internet]. 2012 Jun 28;366(26):2455–65. Available from: http://www.ncbi.nlm.nih.gov/pubmed/22658128
  84. 84. 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. J Clin Oncol [Internet]. 2015 Dec 1;33(34):4015–22. Available from: http://www.ncbi.nlm.nih.gov/pubmed/26351349
  85. 85. Varga A, Piha-Paul SA, Ott PA, Mehnert JM, Berton-Rigaud D, Johnson EA, Cheng JD, Yuan S, Rubin EH. Antitumor activity and safety of pembrolizumab in patients (pts) with PD-L1 positive advanced ovarian cancer: interim results from a phase Ib study. J Clin Oncol. 2015;33(suppl; abstr 5509).
  86. 86. Birkbak NJ, Kochupurakkal B, Izarzugaza JMG, Eklund AC, Li Y, Liu J, et al. Tumor mutation burden forecasts outcome in ovarian cancer with BRCA1 or BRCA2 mutations. PLoS One [Internet]. 2013;8(11):e80023. Available from: http://www.ncbi.nlm.nih.gov/pubmed/24265793
  87. 87. Ku GY, Yuan J, Page DB, Schroeder SEA, Panageas KS, Carvajal RD, et al. Single-institution experience with ipilimumab in advanced melanoma patients in the compassionate use setting: lymphocyte count after 2 doses correlates with survival. Cancer [Internet]. 2010 Apr 1;116(7):1767–75. Available from: http://www.ncbi.nlm.nih.gov/pubmed/20143434
  88. 88. Liakou CI, Kamat A, Tang DN, Chen H, Sun J, Troncoso P, et al. CTLA-4 blockade increases IFNgamma-producing CD4+ICOShi cells to shift the ratio of effector to regulatory T cells in cancer patients. Proc Natl Acad Sci U S A [Internet]. 2008 Sep 30;105(39):14987–92. Available from: http://www.ncbi.nlm.nih.gov/pubmed/18818309
  89. 89. Yuan J, Gnjatic S, Li H, Powel S, Gallardo HF, Ritter E, et al. CTLA-4 blockade enhances polyfunctional NY-ESO-1 specific T cell responses in metastatic melanoma patients with clinical benefit. Proc Natl Acad Sci U S A [Internet]. 2008 Dec 23;105(51):20410–5. Available from: http://www.ncbi.nlm.nih.gov/pubmed/19074257
  90. 90. Hodi FS, Mihm MC, Soiffer RJ, Haluska FG, Butler M, Seiden M V, et al. Biologic activity of cytotoxic T lymphocyte-associated antigen 4 antibody blockade in previously vaccinated metastatic melanoma and ovarian carcinoma patients. Proc Natl Acad Sci U S A [Internet]. 2003 Apr 15;100(8):4712–7. Available from: http://www.ncbi.nlm.nih.gov/pubmed/12682289
  91. 91. Hodi FS, Butler M, Oble DA, Seiden M V, Haluska FG, Kruse A, et al. Immunologic and clinical effects of antibody blockade of cytotoxic T lymphocyte-associated antigen 4 in previously vaccinated cancer patients. Proc Natl Acad Sci U S A [Internet]. 2008 Feb 26;105(8):3005–10. Available from: http://www.ncbi.nlm.nih.gov/pubmed/18287062
  92. 92. Disis ML, Patel MR, Pant S, Hamilton EP, Lockhart AC, Kelly K, Beck JT, Gordon MS, Weiss GJ, Taylor MH, Chaves J, Mita AC, Chin KM, von Heydebreck A, Cuiller J-M. Avelumab (MSB0010718C; anti-PD-L1) in patients with recurrent/refractory ovarian cancer from the JAVELIN Solid Tumor phase Ib trial: safety and clinical activity. J Clin Oncol. 2016;34(suppl; abstr 5533).
  93. 93. Frederick P. Vulvar cancer (squamous cell carcinoma). NCCN Clin Pract Guidel Oncol. Version 1.2017-October 4, 2016; p1–51
  94. 94. Deppe G, Mert I, Belotte J, Winer IS. Chemotherapy of vulvar cancer: a review. Wien Klin Wochenschr [Internet]. 2013 Mar;125(5–6):119–28. Available from: http://www.ncbi.nlm.nih.gov/pubmed/23519539
  95. 95. Reade CJ, Eiriksson LR, Mackay H. Systemic therapy in squamous cell carcinoma of the vulva: current status and future directions. Gynecol Oncol [Internet]. 2014 Mar;132(3):780–9. Available from: http://www.ncbi.nlm.nih.gov/pubmed/24296343
  96. 96. Morris VK, Ciombor KK, Salem ME, Nimeiri HS, Iqbal S, Singh PP, Polite BN, Deming DA, Chan E, Wade JL, Bekaii-Saab TS, Uronis HE, Pasia MG, Bland G, Rober T. NCI9673: a multi-institutional eETCTN phase II study of nivolumab in refractory metastatic squamous cell carcinoma of the anal canal (SCCA). J Clin Oncol. 2016;34(suppl; abstr 3503).
  97. 97. Bray F, Ferlay J, Laversanne M, Brewster DH, Gombe Mbalawa C, Kohler B, et al. Cancer Incidence in Five Continents: Inclusion criteria, highlights from Volume X and the global status of cancer registration. Int J Cancer [Internet]. 2015;137(9):2060–71. Available from: http://www.ncbi.nlm.nih.gov/pubmed/26135522
  98. 98. Koh W-J, Greer BE, Abu-Rustum NR, Apte SM, Campos SM, Chan J, et al. Uterine neoplasms, version 1.2014. J Natl Compr Canc Netw [Internet]. 2014 Feb;12(2):248–80. Available from: http://www.ncbi.nlm.nih.gov/pubmed/24586086
  99. 99. Colombo N, Preti E, Landoni F, Carinelli S, Colombo A, Marini C, et al. Endometrial cancer: ESMO Clinical Practice Guidelines for diagnosis, treatment and follow-up. Ann Oncol [Internet]. 2013;24(suppl 6):vi33–8. Available from: http://www.ncbi.nlm.nih.gov/pubmed/24078661
  100. 100. Palisoul M, Mutch DG. The clinical management of inoperable endometrial carcinoma. Expert Rev Anticancer Ther [Internet]. 2016 May;16(5):515–21. Available from: http://www.ncbi.nlm.nih.gov/pubmed/26999568
  101. 101. Sorbe B, Andersson H, Boman K, Rosenberg P, Kalling M. Treatment of primary advanced and recurrent endometrial carcinoma with a combination of carboplatin and paclitaxel-long-term follow-up. Int J Gynecol Cancer [Internet]. 18(4):803–8. Available from: http://www.ncbi.nlm.nih.gov/pubmed/17944917
  102. 102. Makker V, Hensley ML, Zhou Q, Iasonos A, Aghajanian CA. Treatment of advanced or recurrent endometrial carcinoma with doxorubicin in patients progressing after paclitaxel/carboplatin: Memorial Sloan-Kettering Cancer Center experience from 1995 to 2009. Int J Gynecol Cancer [Internet]. 2013 Jun;23(5):929–34. Available from: http://www.ncbi.nlm.nih.gov/pubmed/23598889
  103. 103. Wadler S, Levy DE, Lincoln ST, Soori GS, Schink JC, Goldberg G. Topotecan is an active agent in the first-line treatment of metastatic or recurrent endometrial carcinoma: Eastern Cooperative Oncology Group Study E3E93. J Clin Oncol [Internet]. 2003 Jun 1;21(11):2110–4. Available from: http://www.ncbi.nlm.nih.gov/pubmed/12775736
  104. 104. Konecny GE, Finkler N, Garcia AA, Lorusso D, Lee PS, Rocconi RP, et al. Second-line dovitinib (TKI258) in patients with FGFR2-mutated or FGFR2-non-mutated advanced or metastatic endometrial cancer: a non-randomised, open-label, two-group, two-stage, phase 2 study. Lancet Oncol [Internet]. 2015;16(6):686–94. Available from: http://www.ncbi.nlm.nih.gov/pubmed/25981814
  105. 105. Felix AS, Sherman ME, Hewitt SM, Gunja MZ, Yang HP, Cora RL, et al. Cell-cycle protein expression in a population-based study of ovarian and endometrial cancers. Front Oncol [Internet]. 2015;5:25. Available from: http://www.ncbi.nlm.nih.gov/pubmed/25709969
  106. 106. Gargiulo P, Della Pepa C, Berardi S, Califano D, Scala S, Buonaguro L, et al. Tumor genotype and immune microenvironment in POLE-ultramutated and MSI-hypermutated endometrial cancers: new candidates for checkpoint blockade immunotherapy? Cancer Treat Rev [Internet]. 2016 Jul;48:61–8. Available from: http://www.ncbi.nlm.nih.gov/pubmed/27362548
  107. 107. Cancer Genome Atlas Research Network, Kandoth C, Schultz N, Cherniack AD, Akbani R, Liu Y, et al. Integrated genomic characterization of endometrial carcinoma. Nature [Internet]. 2013 May 2;497(7447):67–73. Available from: http://www.ncbi.nlm.nih.gov/pubmed/23636398
  108. 108. 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 Res [Internet]. 2013 Dec;33(12):5495–500. Available from: http://www.ncbi.nlm.nih.gov/pubmed/24324087
  109. 109. Coosemans A, Tuyaerts S, Vanderstraeten A, Vergote I, Amant F, Van Gool SW. Dendritic cell immunotherapy in uterine cancer. Hum Vaccin Immunother [Internet]. 2014;10(7):1822–7. Available from: http://www.ncbi.nlm.nih.gov/pubmed/25424788
  110. 110. Coosemans A, Vanderstraeten A, Tuyaerts S, Verschuere T, Moerman P, Berneman Z, et al. Immunological response after WT1 mRNA-loaded dendritic cell immunotherapy in ovarian carcinoma and carcinosarcoma. Anticancer Res [Internet]. 2013 Sep;33(9):3855–9. Available from: http://www.ncbi.nlm.nih.gov/pubmed/24023319
  111. 111. Le DT, Uram JN, Wang H, Bartlett BR, Kemberling H, Eyring AD, et al. PD-1 blockade in tumors with mismatch-repair deficiency. N Engl J Med [Internet]. 2015 Jun 25;372(26):2509–20. Available from: http://www.ncbi.nlm.nih.gov/pubmed/26028255

Written By

Marcus Vetter and Viola Heinzelmann-Schwarz

Submitted: 26 May 2016 Reviewed: 25 January 2017 Published: 26 April 2017