Skip to main content
American Journal of Clinical and Experimental Urology logoLink to American Journal of Clinical and Experimental Urology
. 2019 Apr 25;7(2):64–79.

Immune reaction by cytoreductive prostatectomy

Geun Taek Lee 1, Arnav Srivastava 1, Young Suk Kwon 1, Isaac Yi Kim 1
PMCID: PMC6526355  PMID: 31139701

Abstract

Prostate cancer (PCa) is the most common non-cutaneous cancer among men and the second leading cause of male cancer deaths in the United States. With no effective cure for advanced disease, the survival rates of castration-resistant disease and metastatic disease remains poor. Treatment via hormonal manipulation, immunotherapy, and chemotherapy remain marginally effective, indicating the need for novel treatment strategies. Cytoreductive prostatectomy (CRP) has grown as a treatment modality for metastatic castration resistant prostate cancer (mCRPC) and an emerging body of literature has demonstrated its survival benefits. In this review, we hope to further explore immunologic changes after CRP and the resultant effects on oncologic outcomes. Conclusively, the data and technical considerations of CRS evolve, CRS may continue to expand treat various type of metastatic cancer. Still, there are little reports about immunological changed after CRP. However, based on technical improvement, CRP and combinational immunotherapy are developing treatments of metastatic disease.

Keywords: Prostate cancer, radical prostatectomy, metastasis, cytoreductive

Introduction

Prostate cancer (PCa) is the most common non-cutaneous cancer among men and the second leading cause of male cancer deaths in the United States [1]. In 2018, it is estimated that 29,430 men will die from PCa. Although radiotherapy and surgery effectively treat localized disease, approximately 30% eventually recur following a definitive therapy. Furthermore, with no effective cure for metastatic disease, the 5-year-survival rate for metastatic PCa is only 29% [1]. In patients with metastatic disease, medical or surgical castration is generally accepted as first-line therapy. Androgen deprivation therapy (ADT) is the cornerstone in the treatment of metastatic PCa, followed by and combined with second-line hormonal treatments such as enzalutamide or abiraterone. However, castration-resistant prostate cancer (CRPC) eventually emerges with a median time of 18-24 months [2-4]. Once CRPC develops, secondary hormonal manipulation, immunotherapy, and chemotherapy are marginally effective and the average life expectancy is approximately 5 years [5,6].

In 1995, Hellman and Weichselbaum [7] introduced the concept of cytoreductive surgery (CRS) - the resection of the primary tumor or metastases to reduce tumor burden-in oligometastatic cancer. More recently, cytoreductive prostatectomy has been explored in the literature. Satkunasivam et al., using the SEER-Medicare database, identified patients ≥66 years old with metastatic PCa treated with radical prostatectomy, intensity modulated radiation therapy, conformal radiation therapy or no local therapy [8]. On multivariable analysis CRP correlated to a 52% decrease (HR 0.48, 95% CI (0.27-0.85), P=0.001) in cancer-specific mortality (CSM), adjusting for socioeconomics, primary tumor characteristics, comorbidities, ADT, and bone radiation within 6 months of diagnosis. Furthermore, a propensity score adjusted analysis, demonstrated a 45% decrease (HR 0.55, 95% CI 0.30-0.72) in CRP relative to no local therapy (NLT) group. Heidenreich et al. in 2015, showed the benefits of CRS in well selected men with metastatic prostate cancer who respond well to neoadjuvant androgen deprivation therapy [9]. A total 23 patients with biopsy-proven prostate cancer (minimal osseous metastases, absence of visceral or extensive lymph node metastases and PSA decrease to <1.0 ng/ml after neoadjuvant ADT) who underwent CRP were analyzed compared with control group consisted of 38 men who were treated with ADT without local therapy. Median time to castration resistant prostate cancer was 40 months (IQR=9-65) and 29 months (IQR=16-59) in the CRP treatment group and control group, respectively (P=0.04). While overall survival was similar in both groups, patients undergoing CRP experienced significantly better clinical progression-free survival (38.6 vs. 26.5 months, P=0.03) and cancer specific survival rates (95.6% vs. 84.2%, P=0.04). Regarding the safety and feasibility of CRP, Sooriakumaran et al. in 2016, published a series, n=106, with newly diagnosed mPCa who underwent CRS [10]. 79.2% of patients had no complications; separately, positive-margin (53.8%), lymphocele (8.5%), and wound infection (4.7%) rates were observed and the authors concluded that CRP is a safe procedure for many. No differences in perioperative complications were seen in M1b relative to M1a patients. In a study by Gandaglia et al. in 2017 [11], perioperative and long-term oncologic outcomes of CRP was found to have an acceptable safety profile no perioperative mortalities. Leyh-Bannurah et al. in 2017, demonstrated CRP had a strong protective effect on patients with one or less risk factor (Subhazard ratio (SHR) 0.16, 95% CI, 0.09-0.28) [12]. In patients with two or more risks, this protective effect reduced but CRP was still effective at decreasing CSM relative to patients with NLT control group (SHR 0.6). In 2017, multicentric prospective data from the Local Treatment of Metastatic Prostate Cancer (LoMP) Trial has also been published [13]. The patients who had at least one histologically confirmed metastatic lesion, excluding pelvic lymphadenopathy were selected. CRP was performed in asymptomatic patients, with resectable primary tumor (n=17). Surgical complications within 3 months postoperatively were analyzed compared with patients who got only standard of care (n=29). In 23.5% of CRP patients, PSA was sustained without death or development of castrate resistance and 44.8% of patients in the NLT, or standard of care group, became castrate resistant and 24.1% of patients died. There was a significant survival benefit for patients undergoing CRP (2-yr OS 100% vs. 55% and 2-yr CSS 100% vs. 61%).

Metastatic prostate cancer immunotherapy

PROSTVAC

PSA-TRICOM (PROSTVAC) is a poxvirus-based peptide vaccine encoding PSA and B7.1, ICAM-1, and LFA-3 (TRICOM). These TRICOM molecules engage with naïve T cells during antigen presentation to enhance the downstream activity of Th1, such as antigen-specific cytotoxic T-cell proliferation [14]. PROSTVAC is well tolerated [15,16] and increases the avidity of cytotoxic T-cell against malignant cells [17]. A randomized phase II, placebo-controlled study enrolling 125 patients with mCRPC presented a median OS was 25.1 months for patients given PROSTVAC vs. 16.6 months for controls (HR=0.56; 95% CI: 0.37-0.85; P=0.0061) [18] and there was no difference in progression free survival (PFS) [18].

DCVAC/PCa

DCVAC/PCa is an autologous dendritic cell (DC) vaccine derived from peripheral blood mononuclear cells. The dendritic cells are obtained by apheresis, pulsed with killed prostate cancer cells, and then-after maturation-injected subcutaneously Podrazil et al. in 2015 published that DCVAC/PCa immunotherapy combined with chemotherapy in patients with mCRPC was well tolerated. Patients were also given 50 mg daily for 1 week oral cyclophosphamide to deplete regulatory T cells [19] and a topical Toll-like receptor agonist at the injection site to enhance immune activation. The authors presented that DCVAC/PCa immunotherapy with chemotherapy resulted in a median OS of 19 months. This compared favorably to both the Halabi nomogram-predicted OS of 11.8 months (HR=0.26; 95% CI: 0.13-0.51, P=0.0001) and the MSKCC nomogram-predicted OS of 13 months (HR=0.33; 95% CI: 0.17-0.63, P=0.0008) [20].

ProstAtak

ProstAtak transfers a herpes simplex virus thymidine kinase to tumor cells via an adenoviral vector. Combining ProstAtak with a prodrug creates cytotoxic and immunostimulatory effects [21] that are well tolerated [22]. Rojas-Martinez et al. in 2013 published that ProstAtak treatment had no significant adverse events related to the treatment and no late toxicities after median follow-up of 11.3 years [23]. In this study, no patients developed metastases. After ProstAtak injection, serum PSA levels rose to a peak during week 1-2 and then fell prior to surgery in 8 of 9 patients. The authors concluded that ProstAtak was well tolerated and led to distribution throughout the intra-prostatic tumor with no significant toxicities.

Sipuleucel-T

Sipuleucel-T (Provenge) is an autologous cellular product derived from a patient’s own harvested peripheral blood mononuclear cells. After harvest immature antigen-presenting cells (APCs) via leukapheresis, cells are incubated with a fusion protein (PA2024), which consists of prostatic acid phosphatase (PAP)-expressed in over 95% of PCa- and granulocyte-macrophage colony-stimulating factor GM-CSF) [24]. In the phase III IMPACT trial, among 512 patients with mCRPC, the median overall survival (OS) in patient receiving Sipuleucel-T was 25.8 months, compared to 21.7 months with placebo [25]. Neoadjuvant sipuleucel-T was associated with a 3-fold increase in activated T cells in 57% (95% CI: 39-79) of post-radical prostatectomy (RP) biopsies compared to pretreatment biopsies (P<0.001) [26-28].

Immune checkpoint inhibitors

T-cell activation is a complex process that requires stimulatory signal. T-cell receptor binding to MHC induces specificity to T-cell activation, but additional costimulatory signals are required. The CD28 receptor is constitutively expressed on the surface of T cells and undergoes activation by binding to B7 ligands (B7-1 and B7-2). CD28:B7-1/2 binding lead to proliferation of T cells, increased T-cell survival, and differentiation. Cytotoxic T-lymphocyte-associated protein-4 (CTLA-4) is a CD28 homolog with much higher binding affinity for B7 [29,30]; however, binding of CTLA-4 to B7 does not induce a stimulatory signal [31,32]. This competitive binding can inhibit the costimulatory signal normally provided by CD28:B7 binding [33-35]. The relative amount of CD28:B7 binding versus CTLA-4:B7 binding determines whether a T cell will undergo activation or deactivation [36]. Inhibition of CTLA-4 can shift the immune system balance toward T-cell activation with increased CD28:B7 binding, resulting in rejection of tumors by the host.

In a phase III trial of Kwon et al. in 2014, studying men with mCRPC refractory to docetaxel to receive bone-directed radiotherapy followed by ipilimumab (10 mg/kg) or placebo (n=799). The trial barely failed to meet its primary endpoint of demonstrating an improvement in median OS (11.2 months for ipilimumab versus 10.0 months for placebo; HR=0.85, P=0.053) [37]. However, statistically significant superior median PFS was seen (4.0 versus 3.0 months; HR=0.70; P<0.0001). In the other phase III study of Beer et al. in 2017, 400 chemotherapy-naïve patients with asymptomatic or minimally symptomatic mCRPC and no known visceral metastases were randomized 2:1 to receive up to 4 doses of ipilimumab 10 mg/kg or placebo every 3 weeks, followed by a maintenance dose every 3 months. There was no improvement in OS [38]. However, median PFS again favored ipilimumab (5.6 months versus 3.8 months; HR=0.67; 95% confidence interval (0.55-0.81)) [38]. PFS data and the subgroup analysis in the former study suggest a subset of patients with mCRPC may still benefit from ipilimumab therapy.

Programmed death 1 (PD-1) is a member of the B7/CD28 family of costimulatory receptors. It regulates T-cell activation through binding to its ligands, programmed death ligand 1 (PD-L1) and programmed death ligand 2 (PD-L2) [39]. PD-1 binding inhibits T-cell proliferation and reduces T-cell survival [39]. PD-1 functions primarily in peripheral tissues, where T cells may encounter the immunosuppressive PD-1 ligands PD-L1 (B7-H1) and PD-L2 (B7-DC), which are expressed by tumor cells, stromal cells, or both [40-43]. PD-1 is primarily expressed on activated T cells and pro-B cells; whereas, PD-L1 is expressed on T cells, APCs, vascular endothelial cells, stromal cells, and cancer cells [44,45]. Inhibition of the interaction between PD-1 and PD-L1 can enhance T-cell responses in vitro and mediate preclinical antitumor activity [42,46]. Small data sets from studies of PD-1/PD-L1 inhibition in prostate cancer have revealed modest activity [47] and confirmed PD-L1 expression in some [48,49] but not all [50] tumor specimens. As single agents, immune checkpoint inhibitors have failed to substantially improve clinical outcomes in prostate cancer [47,48,50]. Increased expression of PD-L1 on circulating DCs has been observed in patients with enzalutamide-resistant prostate cancer, suggesting a role for anti-PD-1/PD-L1 treatment in castration-resistant disease [48].

When tumor antigen specific T cells recognize their antigen, signaling through the T cell receptor (TCR) induces interferons (IFNs) [51] and simultaneously express activation-induced regulatory receptors including PD-1 [52]. The IFNs amplify the immune response and attracting other leukocytes such as natural killer (NK) cells and macrophages [53]. However, IFNs also increase the expression of IFN-inducible immune suppressive factors such as PD-L1 and indolamine 2, 3 dioxygenase [54,55]. This is an adaptive process immune resistance that inhibits immune and inflammatory responses, and cancer uses this phenomenon to protect the tumor’s growth. Frequently, cancer cells escape immune system and activate tumor growth using immunosuppressive mechanisms. Strongly or chronically activated T cells express more PD-1, and most cells express its ligand PD-L1 after exposure to T cell-derived IFNγ. In adaptive immune resistance process, these T cell response will increase suppression of activated CD8+ T cells [56].

While checkpoint inhibitor monotherapies have not substantially improved clinical outcomes for patients with prostate cancer, approaches that combine immune checkpoint inhibitors, vaccines, and other agents are under investigation to manipulate the immune system leading to increased antitumor activity.

Combinational immunotherapy

Even though there are many trials studying single agent immunotherapy, it has thus far demonstrated only modest survival benefit to mPCa. Consequently, new research efforts have focused on combination immunotherapy. For example, a completed phase I clinical trial was tested sipuleucel-T in combination with ipilimumab (NCT01832870) and an ongoing phase II trial testing this combination with sequence variation is recruiting subjects (NCT01804465). Two phase II studies were completed sipuleucel-T with enzalutamide in mCRPC (NCT01981122) and concurrent vs. sequential sipuleucel-T and abiraterone treatment in mCRPC (NCT01487863). A phase II study testing PROSTVAC with or without ipilimumab in the neoadjuvant setting is currently recruiting patients (NCT02506114). A phase I/II study currently underway combines a DNA vaccine encoding PAP plus pembrolizumab in sequence or concurrently in mCRPC patients (NCT02499835, NCT03248570, NCT02861573, and NCT02499835). A phase I/II study currently recruiting combines PROSTVAC with ipilimumab, nivolumab, or the triple combination (NCT02933255).

Pro-tumorigenic immune change by cancer and the effect of cytoreductive surgery

Thus far the literature examining changes to the immune system after cytoreductive prostate surgery remains sparse. As such we will review the effects of CRS among other cancers to provide insight into the effects of CRP.

Host immune response to various cancer has a strong influence on clinical outcomes [57,58]. In particular, the presence of CD8+ tumor-infiltrating lymphocytes (TIL) is associated with prolonged PFS and overall survival [59-63]. Additionally, other features of cytolytic CD8+ T cell responses are also positively associated with survival [58]. CD8+ and CD20+ TIL showed the strongest association with survival [63]. In ovarian cancer, 73.1% (79/108) of tumors were positive for intraepithelial CD8+ TIL and 19.4% (20/103) contained intraepithelial CD20+ TIL [64]. Absolute lymphocyte count (ALC) and TIL are independent immunological parameters associated with outcome in high-grade serous carcinoma (HGSC). In the case of absolute lymphocyte count (ALC), this appears to reflect an association with disease burden rather than an immunological mechanism [64]. However, simply increasing ALC may not be sufficient to promote clinically significant antitumor responses. Several markers of inflammation have been associated with increased tumor burden and/or adverse outcome in ovarian cancer, including high neutrophil-to-lymphocyte ratio; high monocyte count [65]; elevated C-reactive protein and hypoalbuminaemia [66] and elevated IL-6 and IL-8 levels.

Helper T cells and cytotoxic T cells change by cancer

Type 1 T helper (Th1) and type 2 T helper (Th2) cells represent two polarized forms of the CD4+ Th cell-mediated specific immune responses. Th1-dominated immune responses predominantly produce a phagocyte-dependent inflammation [67,68]. Type 2 Th (Th2) cells secrete many cytokines such as IL4, IL5, IL6, IL9, IL10, and IL13, which inhibit several functions of phagocytic cells [69-72]. Decker et al. in 1996 and Ishikawa et al. in 2009 presented that changes in the ratio of Th1/Th2 CD4+ cells peak around 2-3 days following surgery, and return to normal levels by post-operative day 14 [73,74]. Surgical stress induces a shift in the Th1/Th2 balance toward Th2, suggesting that cell-mediated immunity is down-regulated and antibody-mediated immunity is up-regulated after surgery.

T cells that secrete cytokines such as interferon gamma (IFN-γ) generate acute inflammation that results in expansion of cytotoxic T cells (CTLs). CTLs are a functional subset of the antigen specific immune response. The key immune cells for antitumor activity are the CTLs which directed against tumor cells susceptible to cell lysis [75]. The presence of antitumor CTLs is a prerequisite for the immune system to attack cancer cells. Immunomodulatory agents attempt to increase the efficacy of CTL activity. In 2003, Zhang et al. published that infiltration of CD3+, CD8+ cytotoxic T cells (CTL) is associated with a better prognosis for ovarian cancer patients [57,59]. In colorectal cancer, it was reported that the T cell infiltration was good marker to predict disease free and OS more accurately compare with the standard TNM staging system [76,77]. Tumor with highly infiltrated T cells and T cell with few T cells were identified retrospectively following CRS. The authors presented that a high T cells infiltrate in colorectal tumors predicts a good outcome of conventional therapy. In the head and neck squamous cell carcinoma (HNSCC), several investigators also published that high numbers of activated CD8+ effector T cells correlates with better survival [78-82]. On the other hand, a greater number of regulatory T cells (Tregs) in tumor infiltrating lymphocyte (TIL) have correlated with a poor prognosis [78-80].

Anti-tumorigenic effect of B-cells

B-cells mainly infiltrated lymphoid structures in the stroma of HGSC metastases. There was a strong B-cell memory response directed at a restricted repertoire of antigens. A positive role for B-cells in the antitumor response was also supported by B-cell depletion in a syngeneic mouse model of peritoneal metastasis. The authors showed that B-cells infiltrating HGSC omental metastases support the development of an antitumor response [83]. In preclinical models of melanoma, squamous cell carcinoma and carcinogen-induced skin cancer, B-cells promote tumor progression through the production of immune regulatory cytokines and immune complexes [84-86]. On the other hand, in human primary tumors, the presence of B-cells in association with tertiary lymphoid structures (TLS) in non-small cell lung carcinoma (NSCLC) and colorectal, ovarian, and pancreatic cancers has been associated with a better prognosis [87-89]. In primary ovarian cancer biopsies, intra-tumor infiltration of CD27-atypical memory B-cells, together with CD8+ T cells, is linked to better prognosis [90]. A very recent study also showed that a high infiltrate of B-cells in primary tumors is linked to the presence of TLS in the microenvironment and improved survival of patients [91]. Montfort et al. in 2017 studied that there was a strong B-cell memory response directed at a restricted repertoire of antigens and production of tumor-specific IgGs by plasma cells in 92 high-grade serous ovarian cancer (HGSOC). In that paper, B-cells were located mainly in lymphoid aggregates, which displayed characteristic features of TLS. There was no significant difference in the number of B-cells in pre and post NACT biopsies, suggesting that chemotherapy neither impairs nor increases B-cell infiltration in omental metastases. B-cells in HGSOC omentum are activated, differentiate to a memory B-cell phenotype and undergo clonal selection. B-cells are implicated in the recruitment of both DCs and granulocytes in omental metastases and more specifically in TLS. Human metastases of HGSOC, B-cells develop memory responses in the tumor microenvironment, likely via their association with TLS. In lung cancer, the presence of B-cell-rich TLS and DCs has been linked to good prognosis and the development of a Th1 signature [92]. Because activated DCs can stimulate CD8+ T cells and promote a cytotoxic response and B cells and DC densities were correlated in HGSOC metastases.

In early studies using murine models of B-cell deficiency, it was suggested that B-cells may actually inhibit the antitumor effect of tumor infiltrating T-cells [93,94]. As a potential mechanism, B-cells have been demonstrated to inhibit the priming effect of CD4+ T-cells on CD8+ T-cells [95]. Yang et al. presented that OS in patients undergoing primary and secondary debulking procedures was 160.6 months in those patients with low B-cell expression as compared to 47.3 months in those with high B-cell expression (P=0.0015) [96]. A greater extent of B-cell infiltration in omental tissue appears to correlate with poorer survival. Milne et al. suggests that tumor infiltrating B cells may be a positive prognostic marker [64].

M2 polarization of macrophages by cancer

Macrophages are classically categorized M1 or pro-inflammatory macrophages, included in the responses of type I helper T cells (Th1), or M2 or anti-inflammatory macrophages, involved in Th2-type responses [97]. M1 macrophages play key roles in the pro-inflammatory and antitumor responses and are induced by interferon (IFN) and lipopolysaccharide (LPS). M2 macrophages are activated by IL-4 and IL-13, and are responsible for angiogenesis and may assist tumor progression [98,99]. Until now, the effect of tumor resection on immune system is very controversial. Zhu et al., in 2017 [100], measured tumor infiltrated with macrophages after surgery win glioblastoma patients. To determine whether macrophages were recruited to the tumor-debulking site, specimens were stained for CD68, which is general marker of tumor associated macrophages (TAM). Tumors from the debulking group exhibited a higher percentage of CD68+ cells (50% vs. 10%, debulking and non-debulking, respectively).

Prior studies have demonstrated that tumor-associated macrophage (TAM) infiltration promotes the progression of various malignancies [101-105]. Wan et al. in 2009 presented that the mean density of TAM was significantly higher in ovarian cancer than in benign ovarian lesions (57.7 vs. 25.3 per vision field, P<0.01). The 5-year survival rate was significantly higher in low-density TAM group than in high-density TAM group of ovarian cancer patients (73.3% vs. 41.2%, P=0.01) [106]. Similar results were published that macrophage infiltration is associated with poor prognosis in numerous malignant tumors [107,108]. Lan et al. in 2013 published that expression of M2-polarized macrophages is associated with poor prognosis for advanced epithelial ovarian cancer [109]. No significant difference was observed in survival between patients in high- and the low-CD68 expression groups. In contrast, the PFS rates and OS rates were significantly higher in the low-CD163 expression group than in the high-CD163 expression group, respectively. Significantly improved 3-year PFS (49.8% vs. 11.0%, P<0.001) and OS (77.4% vs. 45.0%, P=0.001) rates in patients in the low-CD163/CD68 ratio group when compared with the high-CD163/CD68 ratio group was also observed. The authors concluded that infiltration of CD163-positive M2 macrophages as well as activation of macrophages towards the M2 phenotype may contribute to poor survival in advanced ovarian cancer. Cornelissen et al. in 2014 studied that ratio of intratumoral macrophage phenotype is a prognostic factor in epithelioid malignant pleural mesothelioma [110]. The number of CD68 and CD163 cells was comparable between the surgery and the non-surgery group, and was no changed in overall survival (OS) in groups. However, the CD163/CD68 ratio significantly correlated with OS in both in the total patient group. The total number of macrophages in tumor tissue did not correlate with OS in both groups, however, the CD163/CD68 ratio correlates with OS in the total patient group.

The primary tumor is believed to orchestrate and maintain the ideal microenvironment for tumor cell proliferation and subsequent metastases and the concept that immunologic intervention would alter the natural course offers unique insights into tumor progression and potential therapeutics.

Immuno-suppressive effect of dendritic cells

Plasmacytoid dendritic cells (pDC) and myeloid dendritic cells (mDC) are main subsets of human dendritic cells (DC) in blood. pDC are identified as a CD4+, CD11c, lineage marker, and HLADR+ cells that express CD123/IL-3 receptor alpha chain and/or as BDCA2- and BDCA4-expressing cells [111]. pDC link innate and adaptive immune responses by promoting the activation and differentiation of natural killer (NK) cells, B cells, myeloid DC (mDC), and T cells [112-114]. Tumor infiltration by pDC may have clinical importance, as underlined by their identification in tumors including melanoma, head and neck, lung, ovarian, and breast cancers [115,116].

In ovarian cancer, several studies have shown an accumulation of pDC in malignant ascites fluid, with decreased blood concentration [115,117,118]. Ascites-originating pDC were shown to favor tumor angiogenesis via the production of TNFα and IL-8 [118]. Labidi-Galy et al. in 2011 presented that ascite pDC were 7.2-fold higher than patient blood pDC (P<0.001) and 13.8-fold higher than TApDC (P<0.001) [119]. mDC were present in malignant ascites but in lower proportions than pDC (P<0.001) while their presence in tumors was scarce. The authors presented that both TLRL-activated TApDC and ascite pDC were able to induce CD4+ T cell proliferation consistently with the acquisition of a fully mature phenotype. pDC play a proinflammatory role in malignant ascites, whereas they are immunosuppressive in tumors. IFN-a produced primarily from pDC, in addition to having direct anti-tumoral activity [120,121], provides an important signal for T helper precursor differentiation in favor of a T helper type 1 immune response [122]. Zou and colleagues [115,123] showing that pDC isolated from malignant ascites are functional in terms of IFN-a production. The authors detail that the accumulation of pDC in ascites (up to 10-fold higher than in tumors) had no impact on patients’ outcome whereas their presence in tumors was deleterious. Moreover, the authors showed that TApDC were strongly inhibited for their innate response.

In other cancers such as breast and pancreatic cancer, blood pDC and mDC were significantly decreased compared with healthy control groups, consistent with prior studies [124,125].

Cytokines for immune suppression

TGF-β and IL-10, were involved in tumor-induced immunosuppression [126-130]. Transforming growth factor beta (TGF-β) is a multifunctional cytokine, whose myriad of functions include its ability to potently suppress the immune system [131-134]. Interleukin 10 (IL-10) is a suppressive cytokine that is produced and released by immune cells such as macrophages, monocytes and different T-cell subsets, as well as by tumor cells. It down-regulates the macrophages function, inhibits the production of IL-2 and interferon γ (IFN-γ) by Th1-cells, as well as decreases the expression of MHC class I molecule on tumor cells, resulting in the development and promotion of cancer [135-138].

Maeda et al. in 1996 presented that sub-optimal primary surgery leads to unfavorable immunological changes in ovarian cancer patients. On the day of primary cytoreduction and 7 days after, the selected serum immunological parameters were determined in 49 patients with confirmed epithelial ovarian cancer (EOC) [130]. In that paper, the level of immunosuppressive (IL-10 and TGF-β1) and pro-inflammatory (IL-6 and IL-8) cytokines was significantly higher in the group of patients with advanced stage of disease, compared to early stage. The overall survival of patients who underwent optimal cytoreduction was significantly higher than that in whom only sub-optimal surgery was performed. Primary cytoreduction resulted in the decrease of both cytokines serum amount. However, in EOC patients with advanced stage of disease the TGF-β1 concentration, despite being lower, still remained intensified. It evidence that primary cytoreduction only partially reduced tumor-induced immunosuppression. Napoletano et al. (2010) [139] also claimed that solely IL-10 serum level decreased after surgery. Both optimal and suboptimal primary surgery weakened systemic immunosuppression in patients with stage III/IV through reduction of IL-10 and TGF-β1 serum level. However, it should be strongly underlined that the retained concentration of TGF-β1 in the serum of patients in whom only sub-optimal cytoreduction was performed was significantly higher in compare to its level in patients operated optimally. In contrast, we and others [140] noticed that there was no significant difference between the IL-10 level in relation to presence or absence of post-operative residual tumor. TGF-β or IL-10 can favor tumor growth and high immunosuppressive effects on T-cell mediated immunity [137,141], as well as is considered to favor tumor growth and metastasis and its high concentration in tumor micro-environment correlates with poor patient outcome [142]. Li et al. in 2010 published that increase number of such regulatory T cells is associated with elevated concentration of TGF-β1 observed in patients in whom only sub-optimal operation was performed. TGFβ is known to convert CD4+CD25- T cells to CD4+CD25+ T cells that maintains peripheral immune tolerance [143-146]. Moreover, Treg, observed in tumor microenvironment of EOC patients, therefore its high number in circulation could facilitate the creation of favorable environment for cancer cells [147]. Since partial removal of tumor mass did not reverse the systemic immunosuppression we can assume that high serum level of TGF-β1 accompanied with enhanced number of CD4+CD25+high cells in advanced EOC patients might contribute to the induction of T-cell suppression, thus allowing the metastases growth from postoperative residual tumor tissue. Similar results was published by Shashikant et al. in 2009 that sub-optimal cytoreduction resulted in unfavorable increase in the percentage of CD4+CD25+high cells in peripheral blood as well as in insufficient decrease in serum concentration of TGF-β. However, the optimal cytoreduction could promote opportunity of ovarian cancer patients in III/IV stage of disease to improve PFS and OS [148].

Interleukin 6 (IL-6) signaling is generally considered pro-tumorigenic cytokine in various malignances [149-152]. However, Tempfer et al. in 1997 and Nowak et al. in 2010 presented that the serum concentration of IL-6 was markedly higher, mainly in EOC patients with advanced stage of the disease [153,154] and primary cytoreduction both optimal and sub-optimal had no influence on its level.

The primary cytoreduction reduced the serum concentration of IL-8 of patients with FIGO stage III/IV, although sub-optimal surgical operation resulted in still high amount of it. Interleukin 8, is a pro-inflammatory cytokine and its overexpression in tumor microenvironment of ovarian cancer patients significantly correlates with disease severity and poor prognosis [155].

Conclusions

CRS has been explored in multiple tumor types [156-162]. Kidney cancer provides one of the most striking examples of the benefits CRS, as cytoreductive nephrectomy plus immunotherapy has previously demonstrated survival improvement [163-167]. However, as data and technical improvements in CRS as well as the role of immunotherapy continue to evolve, we may see expansion in CRS. In this paper, we overviewed about immunological change in the context of interaction of tumor and tumor microenvironment and the effect of CRS in immune system. The interaction between tumor and microenvironment such as immune cells derived to pro-tumorigenic such as M2 macrophages and Th2 cells and increased immunosuppressive cytokines such as TGF-β and IL-10. Therefore, we expected that CRS may decrease these driving forces and the combination with CRP and immunotherapy or chemotherapy may help to survival benefit. Still, there are little reports about immunological changed after CRP. If many ongoing clinical trial data analysis are accumulated, it can show the right way about CRP.

Acknowledgements

This work was supported by the Prostate Cancer Research Program Idea Development Award for Established Investigators from the United States Department of Defense Office of the Congressionally Directed Medical Research Program (W81XWH-17-1-0359), and the generous support from the Marion and Norman Tanzman Charitable Foundation and Mr. Malcolm Wernik.

Disclosure of conflict of interest

None.

References

  • 1.Siegel RL, Miller KD, Jemal A. Cancer statistics, 2018. CA Cancer J Clin. 2018;68:7–30. doi: 10.3322/caac.21442. [DOI] [PubMed] [Google Scholar]
  • 2.Schellhammer P, Sharifi R, Block N, Soloway M, Venner P, Patterson AL, Sarosdy M, Vogelzang N, Jones J, Kolvenbag G. Maximal androgen blockade for patients with metastatic prostate cancer: outcome of a controlled trial of bicalutamide versus flutamide, each in combination with luteinizing hormone-releasing hormone analogue therapy. Casodex Combination Study Group. Urology. 1996;47(Suppl):54–60. doi: 10.1016/s0090-4295(96)80010-0. discussion 80-4. [DOI] [PubMed] [Google Scholar]
  • 3.Crawford ED, Eisenberger MA, McLeod DG, Spaulding JT, Benson R, Dorr FA, Blumenstein BA, Davis MA, Goodman PJ. A controlled trial of leuprolide with and without flutamide in prostatic carcinoma. N Engl J Med. 1989;321:419–424. doi: 10.1056/NEJM198908173210702. [DOI] [PubMed] [Google Scholar]
  • 4.Caubet JF, Tosteson TD, Dong EW, Naylon EM, Whiting GW, Ernstoff MS, Ross SD. Maximum androgen blockade in advanced prostate cancer: a meta-analysis of published randomized controlled trials using nonsteroidal antiandrogens. Urology. 1997;49:71–78. doi: 10.1016/S0090-4295(96)00325-1. [DOI] [PubMed] [Google Scholar]
  • 5.Debes JD, Tindall DJ. Mechanisms of androgen-refractory prostate cancer. N Engl J Med. 2004;351:1488–1490. doi: 10.1056/NEJMp048178. [DOI] [PubMed] [Google Scholar]
  • 6.Feldman BJ, Feldman D. The development of androgen-independent prostate cancer. Nat Rev Cancer. 2001;1:34–45. doi: 10.1038/35094009. [DOI] [PubMed] [Google Scholar]
  • 7.Hellman S, Weichselbaum RR. Oligometastases. J. Clin. Oncol. 1995;13:8–10. doi: 10.1200/JCO.1995.13.1.8. [DOI] [PubMed] [Google Scholar]
  • 8.Satkunasivam R, Kim AE, Desai M, Nguyen MM, Quinn DI, Ballas L, Lewinger JP, Stern MC, Hamilton AS, Aron M, Gill IS. Radical prostatectomy or external beam radiation therapy vs no local therapy for survival benefit in metastatic prostate cancer: a SEER-medicare analysis. J Urol. 2015;194:378–385. doi: 10.1016/j.juro.2015.02.084. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 9.Heidenreich A, Pfister D, Porres D. Cytoreductive radical prostatectomy in patients with prostate cancer and low volume skeletal metastases: results of a feasibility and case-control study. J Urol. 2015;193:832–838. doi: 10.1016/j.juro.2014.09.089. [DOI] [PubMed] [Google Scholar]
  • 10.Sooriakumaran P, Karnes J, Stief C, Copsey B, Montorsi F, Hammerer P, Beyer B, Moschini M, Gratzke C, Steuber T, Suardi N, Briganti A, Manka L, Nyberg T, Dutton SJ, Wiklund P, Graefen M. A multi-institutional analysis of perioperative outcomes in 106 men who underwent radical prostatectomy for distant metastatic prostate cancer at presentation. Eur Urol. 2016;69:788–794. doi: 10.1016/j.eururo.2015.05.023. [DOI] [PubMed] [Google Scholar]
  • 11.Gandaglia G, Fossati N, Stabile A, Bandini M, Rigatti P, Montorsi F, Briganti A. Radical prostatectomy in men with oligometastatic prostate cancer: results of a single-institution series with long-term follow-up. Eur Urol. 2017;72:289–292. doi: 10.1016/j.eururo.2016.08.040. [DOI] [PubMed] [Google Scholar]
  • 12.Leyh-Bannurah SR, Gazdovich S, Budäus L, Zaffuto E, Briganti A, Abdollah F, Montorsi F, Schiffmann J, Menon M, Shariat SF, Fisch M, Chun F, Steuber T, Huland H, Graefen M, Karakiewicz P. Local therapy improves survival in metastatic prostate cancer. Eur Urol. 2017;72:118–124. doi: 10.1016/j.eururo.2017.03.020. [DOI] [PubMed] [Google Scholar]
  • 13.Poelaert F, Verbaeys C, Rappe B, Kimpe B, Billiet I, Plancke H, Decaestecker K, Fonteyne V, Buelens S, Lumen N. Cytoreductive prostatectomy for metastatic prostate cancer: first lessons learned from the multicentric prospective local treatment of metastatic prostate cancer (LoMP) trial. Urology. 2017;106:146–152. doi: 10.1016/j.urology.2017.02.051. [DOI] [PubMed] [Google Scholar]
  • 14.Hodge JW, Sabzevari H, Yafal AG, Gritz L, Lorenz MG, Schlom J. A triad of costimulatory molecules synergize to amplify T-cell activation. Cancer Res. 1999;59:5800–5807. [PubMed] [Google Scholar]
  • 15.DiPaola RS, Plante M, Kaufman H, Petrylak DP, Israeli R, Lattime E, Manson K, Schuetz T. A phase I trial of pox PSA vaccines (PROSTVAC-VF) with B7-1, ICAM-1, and LFA-3 co-stimulatory molecules (TRICOM) in patients with prostate cancer. J Transl Med. 2006;4:1. doi: 10.1186/1479-5876-4-1. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 16.Arlen PM, Skarupa L, Pazdur M, Seetharam M, Tsang KY, Grosenbach DW, Feldman J, Poole DJ, Litzinger M, Steinberg SM, Jones E, Chen C, Marte J, Parnes H, Wright J, Dahut W, Schlom J, Gulley JL. Clinical safety of a viral vector based prostate cancer vaccine strategy. J Urol. 2007;178:1515–1520. doi: 10.1016/j.juro.2007.05.117. [DOI] [PubMed] [Google Scholar]
  • 17.Hodge JW, Chakraborty M, Kudo-Saito C, Garnett CT, Schlom J. Multiple costimulatory modalities enhance CTL avidity. J Immunol. 2005;174:5994–6004. doi: 10.4049/jimmunol.174.10.5994. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 18.Kantoff PW, Schuetz TJ, Blumenstein BA, Glode LM, Bilhartz DL, Wyand M, Manson K, Panicali DL, Laus R, Schlom J, Dahut WL, Arlen PM, Gulley JL, Godfrey WR. Overall survival analysis of a phase II randomized controlled trial of a Poxviral-based PSA-targeted immunotherapy in metastatic castration-resistant prostate cancer. J. Clin. Oncol. 2010;28:1099–1105. doi: 10.1200/JCO.2009.25.0597. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 19.Le DT, Jaffee EM. Regulatory T-cell modulation using cyclophosphamide in vaccine approaches: a current perspective. Cancer Res. 2012;72:3439–3444. doi: 10.1158/0008-5472.CAN-11-3912. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 20.Podrazil M, Horvath R, Becht E, Rozkova D, Bilkova P, Sochorova K, Hromadkova H, Kayserova J, Vavrova K, Lastovicka J, Vrabcova P, Kubackova K, Gasova Z, Jarolim L, Babjuk M, Spisek R, Bartunkova J, Fucikova J. Phase I/II clinical trial of dendritic-cell based immunotherapy (DCVAC/PCa) combined with chemotherapy in patients with metastatic, castration-resistant prostate cancer. Oncotarget. 2015;6:18192–18205. doi: 10.18632/oncotarget.4145. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 21.Mesnil M, Yamasaki H. Bystander effect in herpes simplex virus-thymidine kinase/ganciclovir cancer gene therapy: role of gap-junctional intercellular communication. Cancer Res. 2000;60:3989–3999. [PubMed] [Google Scholar]
  • 22.Herman JR, Adler HL, Aguilar-Cordova E, Rojas-Martinez A, Woo S, Timme TL, Wheeler TM, Thompson TC, Scardino PT. In situ gene therapy for adenocarcinoma of the prostate: a phase I clinical trial. Hum Gene Ther. 1999;10:1239–1249. doi: 10.1089/10430349950018229. [DOI] [PubMed] [Google Scholar]
  • 23.Rojas-Martínez A, Manzanera AG, Sukin SW, Esteban-María J, González-Guerrero JF, Gomez-Guerra L, Garza-Guajardo R, Flores-Gutiérrez JP, Elizondo Riojas G, Delgado-Enciso I, Ortiz-López R, Aguilar LK, Butler EB, Barrera-Saldaña HA, Aguilar-Cordova E. Intraprostatic distribution and long-term follow-up after AdV-tk immunotherapy as neoadjuvant to surgery in patients with prostate cancer. Cancer Gene Ther. 2013;20:642–649. doi: 10.1038/cgt.2013.56. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 24.Goldstein NS. Immunophenotypic characterization of 225 prostate adenocarcinomas with intermediate or high gleason scores. Am J Clin Pathol. 2002;117:471–477. doi: 10.1309/G6PR-Y774-X738-FG2K. [DOI] [PubMed] [Google Scholar]
  • 25.Kantoff PW, Higano CS, Shore ND, Berger ER, Small EJ, Penson DF, Redfern CH, Ferrari AC, Dreicer R, Sims RB, Xu Y, Frohlich MW, Schellhammer PF IMPACT Study Investigators. Sipuleucel-T immunotherapy for castration-resistant prostate cancer. N Engl J Med. 2010;363:411–422. doi: 10.1056/NEJMoa1001294. [DOI] [PubMed] [Google Scholar]
  • 26.Sheikh NA, Petrylak D, Kantoff PW, Dela Rosa C, Stewart FP, Kuan LY, Whitmore JB, Trager JB, Poehlein CH, Frohlich MW, Urdal DL. Sipuleucel-T immune parameters correlate with survival: an analysis of the randomized phase 3 clinical trials in men with castration-resistant prostate cancer. Cancer Immunol Immunother. 2013;62:137–47. doi: 10.1007/s00262-012-1317-2. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 27.McNeel DG, Gardner TA, Higano CS, Kantoff PW, Small EJ, Wener MH, Sims RB, DeVries T, Sheikh NA, Dreicer R. A transient increase in eosinophils is associated with prolonged survival in men with metastatic castration-resistant prostate cancer who receive sipuleucel-T. Cancer Immunol Res. 2014;2:988–999. doi: 10.1158/2326-6066.CIR-14-0073. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 28.GuhaThakurta D, Sheikh NA, Fan LQ, Kandadi H, Meagher TC, Hall SJ, Kantoff PW, Higano CS, Small EJ, Gardner TA, Bailey K, Vu T, DeVries T, Whitmore JB, Frohlich MW, Trager JB, Drake CG. Humoral immune response against nontargeted tumor antigens after treatment with sipuleucel-T and its association with improved clinical outcome. Clin Cancer Res. 2015;21:3619–3630. doi: 10.1158/1078-0432.CCR-14-2334. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 29.Chambers CA, Kuhns MS, Egen JG, Allison JP. CTLA-4-mediated inhibition in regulation of T cell responses: mechanisms and manipulation in tumor immunotherapy. Annu Rev Immunol. 2001;19:565–94. doi: 10.1146/annurev.immunol.19.1.565. [DOI] [PubMed] [Google Scholar]
  • 30.Collins AV, Brodie DW, Gilbert RJ, Iaboni A, Manso-Sancho R, Walse B, Stuart DI, van der Merwe PA, Davis SJ. The interaction properties of costimulatory molecules revisited. Immunity. 2002;17:201–210. doi: 10.1016/s1074-7613(02)00362-x. [DOI] [PubMed] [Google Scholar]
  • 31.Alme AK, Karir BS, Faltas BM, Drake CG. Blocking immune checkpoints in prostate, kidney, and urothelial cancer: an overview. Urol Oncol. 2016;34:171–181. doi: 10.1016/j.urolonc.2016.01.006. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 32.Goswami S, Aparicio A, Subudhi SK. Immune checkpoint therapies in prostate cancer. Cancer J. 2016;22:117–120. doi: 10.1097/PPO.0000000000000176. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 33.Chambers JD, Neumann PJ. Listening to provenge-what a costly cancer treatment says about future medicare policy. N Engl J Med. 2011;364:1687–1689. doi: 10.1056/NEJMp1103057. [DOI] [PubMed] [Google Scholar]
  • 34.Egen JG, Kuhns MS, Allison JP. CTLA-4: new insights into its biological function and use in tumor immunotherapy. Nat Immunol. 2002;3:611–618. doi: 10.1038/ni0702-611. [DOI] [PubMed] [Google Scholar]
  • 35.Parry RV, Chemnitz JM, Frauwirth KA, Lanfranco AR, Braunstein I, Kobayashi SV, Linsley PS, Thompson CB, Riley JL. CTLA-4 and PD-1 receptors inhibit T-cell activation by distinct mechanisms. Mol Cell Biol. 2005;25:9543–9553. doi: 10.1128/MCB.25.21.9543-9553.2005. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 36.Krummel MF, Allison JP. CD28 and CTLA-4 have opposing effects on the response of T cells to stimulation. J Exp Med. 1995;182:459–465. doi: 10.1084/jem.182.2.459. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 37.Kwon ED, Drake CG, Scher HI, Fizazi K, Bossi A, van den Eertwegh AJ, Krainer M, Houede N, Santos R, Mahammedi H, Ng S, Maio M, Franke FA, Sundar S, Agarwal N, Bergman AM, Ciuleanu TE, Korbenfeld E, Sengeløv L, Hansen S, Logothetis C, Beer TM, McHenry MB, Gagnier P, Liu D, Gerritsen WR CA184-043 Investigators. Ipilimumab versus placebo after radiotherapy in patients with metastatic castration-resistant prostate cancer that had progressed after docetaxel chemotherapy (CA184-043): a multicentre, randomised, double-blind, phase 3 trial. Lancet Oncol. 2014;15:700–712. doi: 10.1016/S1470-2045(14)70189-5. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 38.Beer TM, Kwon ED, Drake CG, Fizazi K, Logothetis C, Gravis G, Ganju V, Polikoff J, Saad F, Humanski P, Piulats JM, Gonzalez Mella P, Ng SS, Jaeger D, Parnis FX, Franke FA, Puente J, Carvajal R, Sengeløv L, McHenry MB, Varma A, van den Eertwegh AJ, Gerritsen W. Randomized, double-blind, phase iii trial of ipilimumab versus placebo in asymptomatic or minimally symptomatic patients with metastatic chemotherapy-naive castration-resistant prostate cancer. J. Clin. Oncol. 2017;35:40–47. doi: 10.1200/JCO.2016.69.1584. [DOI] [PubMed] [Google Scholar]
  • 39.Keir ME, Butte MJ, Freeman GJ, Sharpe AH. PD-1 and its ligands in tolerance and immunity. Annu Rev Immunol. 2008;26:677–704. doi: 10.1146/annurev.immunol.26.021607.090331. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 40.Dong H, Zhu G, Tamada K, Chen L. B7-H1, a third member of the B7 family, co-stimulates T-cell proliferation and interleukin-10 secretion. Nat Med. 1999;5:1365–1369. doi: 10.1038/70932. [DOI] [PubMed] [Google Scholar]
  • 41.Freeman GJ, Long AJ, Iwai Y, Bourque K, Chernova T, Nishimura H, Fitz LJ, Malenkovich N, Okazaki T, Byrne MC, Horton HF, Fouser L, Carter L, Ling V, Bowman MR, Carreno BM, Collins M, Wood CR, Honjo T. Engagement of the PD-1 immunoinhibitory receptor by a novel B7 family member leads to negative regulation of lymphocyte activation. J Exp Med. 2000;192:1027–1034. doi: 10.1084/jem.192.7.1027. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 42.Dong H, Strome SE, Salomao DR, Tamura H, Hirano F, Flies DB, Roche PC, Lu J, Zhu G, Tamada K, Lennon VA, Celis E, Chen L. Tumor-associated B7-H1 promotes T-cell apoptosis: a potential mechanism of immune evasion. Nat Med. 2002;8:793–800. doi: 10.1038/nm730. [DOI] [PubMed] [Google Scholar]
  • 43.Topalian SL, Drake CG, Pardoll DM. Targeting the PD-1/B7-H1(PD-L1) pathway to activate anti-tumor immunity. Curr Opin Immunol. 2012;24:207–212. doi: 10.1016/j.coi.2011.12.009. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 44.Boussiotis VA. Molecular and biochemical aspects of the PD-1 checkpoint pathway. N Engl J Med. 2016;375:1767–1778. doi: 10.1056/NEJMra1514296. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 45.Ahmadzadeh M, Johnson LA, Heemskerk B, Wunderlich JR, Dudley ME, White DE, Rosenberg SA. Tumor antigen-specific CD8 T cells infiltrating the tumor express high levels of PD-1 and are functionally impaired. Blood. 2009;114:1537–1544. doi: 10.1182/blood-2008-12-195792. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 46.Iwai Y, Ishida M, Tanaka Y, Okazaki T, Honjo T, Minato N. Involvement of PD-L1 on tumor cells in the escape from host immune system and tumor immunotherapy by PD-L1 blockade. Proc Natl Acad Sci U S A. 2002;99:12293–12297. doi: 10.1073/pnas.192461099. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 47.Topalian SL, Hodi FS, Brahmer JR, Gettinger SN, Smith DC, McDermott DF, Powderly JD, Carvajal RD, Sosman JA, Atkins MB, Leming PD, Spigel DR, Antonia SJ, Horn L, Drake CG, Pardoll DM, Chen L, Sharfman WH, Anders RA, Taube JM, McMiller TL, Xu H, Korman AJ, Jure-Kunkel M, Agrawal S, McDonald D, Kollia GD, Gupta A, Wigginton JM, Sznol M. Safety, activity, and immune correlates of anti-PD-1 antibody in cancer. N Engl J Med. 2012;366:2443–2454. doi: 10.1056/NEJMoa1200690. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 48.Bishop JL, Sio A, Angeles A, Roberts ME, Azad AA, Chi KN, Zoubeidi A. PD-L1 is highly expressed in enzalutamide resistant prostate cancer. Oncotarget. 2015;6:234–242. doi: 10.18632/oncotarget.2703. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 49.Graff JN, Alumkal JJ, Drake CG, Thomas GV, Redmond WL, Farhad M, Cetnar JP, Ey FS, Bergan RC, Slottke R, Beer TM. Early evidence of anti-PD-1 activity in enzalutamide-resistant prostate cancer. Oncotarget. 2016;7:52810–52817. doi: 10.18632/oncotarget.10547. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 50.Taube JM, Klein A, Brahmer JR, Xu H, Pan X, Kim JH, Chen L, Pardoll DM, Topalian SL, Anders RA. Association of PD-1, PD-1 ligands, and other features of the tumor immune microenvironment with response to anti-PD-1 therapy. Clin Cancer Res. 2014;20:5064–5074. doi: 10.1158/1078-0432.CCR-13-3271. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 51.Corre B, Perrier J, El Khouri M, Cerboni S, Pellegrini S, Michel F. Type I interferon potentiates T-cell receptor mediated induction of IL-10-producing CD4(+) T cells. Eur J Immunol. 2013;43:2730–2740. doi: 10.1002/eji.201242977. [DOI] [PubMed] [Google Scholar]
  • 52.Ribas A. Adaptive immune resistance: how cancer protects from immune attack. Cancer Discov. 2015;5:915–919. doi: 10.1158/2159-8290.CD-15-0563. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 53.Arango CM, Parra DC, Gómez LF, Lema L, Lobelo F, Ekelund U. Screen time, cardiorespiratory fitness and adiposity among school-age children from Monteria, Colombia. J Sci Med Sport. 2014;17:491–495. doi: 10.1016/j.jsams.2013.09.010. [DOI] [PubMed] [Google Scholar]
  • 54.Rooney MS, Shukla SA, Wu CJ, Getz G, Hacohen N. Molecular and genetic properties of tumors associated with local immune cytolytic activity. Cell. 2015;160:48–61. doi: 10.1016/j.cell.2014.12.033. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 55.Spranger S, Spaapen RM, Zha Y, Williams J, Meng Y, Ha TT, Gajewski TF. Up-regulation of PD-L1, IDO, and T(regs) in the melanoma tumor microenvironment is driven by CD8(+) T cells. Sci Transl Med. 2013;5:200ra116. doi: 10.1126/scitranslmed.3006504. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 56.Tumeh PC, Harview CL, Yearley JH, Shintaku IP, Taylor EJ, Robert L, Chmielowski B, Spasic M, Henry G, Ciobanu V, West AN, Carmona M, Kivork C, Seja E, Cherry G, Gutierrez AJ, Grogan TR, Mateus C, Tomasic G, Glaspy JA, Emerson RO, Robins H, Pierce RH, Elashoff DA, Robert C, Ribas A. PD-1 blockade induces responses by inhibiting adaptive immune resistance. Nature. 2014;515:568–571. doi: 10.1038/nature13954. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 57.Zhang L, Conejo-Garcia JR, Katsaros D, Gimotty PA, Massobrio M, Regnani G, Makrigiannakis A, Gray H, Schlienger K, Liebman MN, Rubin SC, Coukos G. Intratumoral T cells, recurrence, and survival in epithelial ovarian cancer. N Engl J Med. 2003;348:203–213. doi: 10.1056/NEJMoa020177. [DOI] [PubMed] [Google Scholar]
  • 58.Nelson BH. The impact of T-cell immunity on ovarian cancer outcomes. Immunol Rev. 2008;222:101–116. doi: 10.1111/j.1600-065X.2008.00614.x. [DOI] [PubMed] [Google Scholar]
  • 59.Sato E, Olson SH, Ahn J, Bundy B, Nishikawa H, Qian F, Jungbluth AA, Frosina D, Gnjatic S, Ambrosone C, Kepner J, Odunsi T, Ritter G, Lele S, Chen YT, Ohtani H, Old LJ, Odunsi K. Intraepithelial CD8+ tumor-infiltrating lymphocytes and a high CD8+/regulatory T cell ratio are associated with favorable prognosis in ovarian cancer. Proc Natl Acad Sci U S A. 2005;102:18538–18543. doi: 10.1073/pnas.0509182102. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 60.Hamanishi J, Mandai M, Iwasaki M, Okazaki T, Tanaka Y, Yamaguchi K, Higuchi T, Yagi H, Takakura K, Minato N, Honjo T, Fujii S. Programmed cell death 1 ligand 1 and tumor-infiltrating CD8+ T lymphocytes are prognostic factors of human ovarian cancer. Proc Natl Acad Sci U S A. 2007;104:3360–3365. doi: 10.1073/pnas.0611533104. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 61.Clarke B, Tinker AV, Lee CH, Subramanian S, van de Rijn M, Turbin D, Kalloger S, Han G, Ceballos K, Cadungog MG, Huntsman DG, Coukos G, Gilks CB. Intraepithelial T cells and prognosis in ovarian carcinoma: novel associations with stage, tumor type, and BRCA1 loss. Mod Pathol. 2009;22:393–402. doi: 10.1038/modpathol.2008.191. [DOI] [PubMed] [Google Scholar]
  • 62.Leffers N, Gooden MJ, de Jong RA, Hoogeboom BN, ten Hoor KA, Hollema H, Boezen HM, van der Zee AG, Daemen T, Nijman HW. Prognostic significance of tumor-infiltrating T-lymphocytes in primary and metastatic lesions of advanced stage ovarian cancer. Cancer Immunol Immunother. 2009;58:449–459. doi: 10.1007/s00262-008-0583-5. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 63.Milne K, Köbel M, Kalloger SE, Barnes RO, Gao D, Gilks CB, Watson PH, Nelson BH. Systematic analysis of immune infiltrates in high-grade serous ovarian cancer reveals CD20, FoxP3 and TIA-1 as positive prognostic factors. PLoS One. 2009;4:e6412. doi: 10.1371/journal.pone.0006412. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 64.Milne K, Alexander C, Webb JR, Sun W, Dillon K, Kalloger SE, Gilks CB, Clarke B, Köbel M, Nelson BH. Absolute lymphocyte count is associated with survival in ovarian cancer independent of tumor-infiltrating lymphocytes. J Transl Med. 2012;10:33. doi: 10.1186/1479-5876-10-33. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 65.Bishara S, Griffin M, Cargill A, Bali A, Gore ME, Kaye SB, Shepherd JH, Van Trappen PO. Pre-treatment white blood cell subtypes as prognostic indicators in ovarian cancer. Eur J Obstet Gynecol Reprod Biol. 2008;138:71–75. doi: 10.1016/j.ejogrb.2007.05.012. [DOI] [PubMed] [Google Scholar]
  • 66.Sharma R, Hook J, Kumar M, Gabra H. Evaluation of an inflammation-based prognostic score in patients with advanced ovarian cancer. Eur J Cancer. 2008;44:251–256. doi: 10.1016/j.ejca.2007.11.011. [DOI] [PubMed] [Google Scholar]
  • 67.Abbas AK, Murphy KM, Sher A. Functional diversity of helper T lymphocytes. Nature. 1996;383:787–793. doi: 10.1038/383787a0. [DOI] [PubMed] [Google Scholar]
  • 68.Romagnani S. Th1/Th2 cells. Inflamm Bowel Dis. 1999;5:285–294. doi: 10.1097/00054725-199911000-00009. [DOI] [PubMed] [Google Scholar]
  • 69.Romagnani S. T-cell subsets (Th1 versus Th2) Ann Allergy Asthma Immunol. 2000;85:9–18. doi: 10.1016/S1081-1206(10)62426-X. quiz 18, 21. [DOI] [PubMed] [Google Scholar]
  • 70.Young DA, Lowe LD, Booth SS, Whitters MJ, Nicholson L, Kuchroo VK, Collins M. IL-4, IL-10, IL-13, and TGF-beta from an altered peptide ligand-specific Th2 cell clone down-regulate adoptive transfer of experimental autoimmune encephalomyelitis. J Immunol. 2000;164:3563–3572. doi: 10.4049/jimmunol.164.7.3563. [DOI] [PubMed] [Google Scholar]
  • 71.McKenzie GJ, Emson CL, Bell SE, Anderson S, Fallon P, Zurawski G, Murray R, Grencis R, McKenzie AN. Impaired development of Th2 cells in IL-13-deficient mice. Immunity. 1998;9:423–432. doi: 10.1016/s1074-7613(00)80625-1. [DOI] [PubMed] [Google Scholar]
  • 72.Rowe J, Macaubas C, Monger TM, Holt BJ, Harvey J, Poolman JT, Sly PD, Holt PG. Antigen-specific responses to diphtheria-tetanus-acellular pertussis vaccine in human infants are initially Th2 polarized. Infect Immun. 2000;68:3873–3877. doi: 10.1128/iai.68.7.3873-3877.2000. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 73.Decker D, Schondorf M, Bidlingmaier F, Hirner A, von Ruecker AA. Surgical stress induces a shift in the type-1/type-2 T-helper cell balance, suggesting down-regulation of cell-mediated and up-regulation of antibody-mediated immunity commensurate to the trauma. Surgery. 1996;119:316–325. doi: 10.1016/s0039-6060(96)80118-8. [DOI] [PubMed] [Google Scholar]
  • 74.Ishikawa M, Nishioka M, Hanaki N, Miyauchi T, Kashiwagi Y, Ioki H, Kagawa A, Nakamura Y. Perioperative immune responses in cancer patients undergoing digestive surgeries. World J Surg Oncol. 2009;7:7. doi: 10.1186/1477-7819-7-7. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 75.Chouaib S. Integrating the quality of the cytotoxic response and tumor susceptibility into the design of protective vaccines in tumor immunotherapy. J Clin Invest. 2003;111:595–597. doi: 10.1172/JCI18044. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 76.Pagès F, Berger A, Camus M, Sanchez-Cabo F, Costes A, Molidor R, Mlecnik B, Kirilovsky A, Nilsson M, Damotte D, Meatchi T, Bruneval P, Cugnenc PH, Trajanoski Z, Fridman WH, Galon J. Effector memory T cells, early metastasis, and survival in colorectal cancer. N Engl J Med. 2005;353:2654–2666. doi: 10.1056/NEJMoa051424. [DOI] [PubMed] [Google Scholar]
  • 77.Galon J, Costes A, Sanchez-Cabo F, Kirilovsky A, Mlecnik B, Lagorce-Pagès C, Tosolini M, Camus M, Berger A, Wind P, Zinzindohoué F, Bruneval P, Cugnenc PH, Trajanoski Z, Fridman WH, Pagès F. Type, density, and location of immune cells within human colorectal tumors predict clinical outcome. Science. 2006;313:1960–1964. doi: 10.1126/science.1129139. [DOI] [PubMed] [Google Scholar]
  • 78.Wansom D, Light E, Worden F, Prince M, Urba S, Chepeha DB, Cordell K, Eisbruch A, Taylor J, D’Silva N, Moyer J, Bradford CR, Kurnit D, Kumar B, Carey TE, Wolf GT. Correlation of cellular immunity with human papillomavirus 16 status and outcome in patients with advanced oropharyngeal cancer. Arch Otolaryngol Head Neck Surg. 2010;136:1267–1273. doi: 10.1001/archoto.2010.211. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 79.Green VL, Michno A, Stafford ND, Greenman J. Increased prevalence of tumour infiltrating immune cells in oropharyngeal tumours in comparison to other subsites: relationship to peripheral immunity. Cancer Immunol Immunother. 2013;62:863–873. doi: 10.1007/s00262-013-1395-9. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 80.Ward MJ, Thirdborough SM, Mellows T, Riley C, Harris S, Suchak K, Webb A, Hampton C, Patel NN, Randall CJ, Cox HJ, Jogai S, Primrose J, Piper K, Ottensmeier CH, King EV, Thomas GJ. Tumour-infiltrating lymphocytes predict for outcome in HPV-positive oropharyngeal cancer. Br J Cancer. 2014;110:489–500. doi: 10.1038/bjc.2013.639. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 81.Wolf GT, Chepeha DB, Bellile E, Nguyen A, Thomas D, McHugh J University of Michigan Head and Neck SPORE Program. Tumor infiltrating lymphocytes (TIL) and prognosis in oral cavity squamous carcinoma: a preliminary study. Oral Oncol. 2015;51:90–95. doi: 10.1016/j.oraloncology.2014.09.006. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 82.Nguyen N, Bellile E, Thomas D, McHugh J, Rozek L, Virani S, Peterson L, Carey TE, Walline H, Moyer J, Spector M, Perim D, Prince M, McLean S, Bradford CR, Taylor JM, Wolf GT Head and Neck SPORE Program Investigators. Tumor infiltrating lymphocytes and survival in patients with head and neck squamous cell carcinoma. Head Neck. 2016;38:1074–1084. doi: 10.1002/hed.24406. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 83.Montfort A, Pearce O, Maniati E, Vincent BG, Bixby L, Böhm S, Dowe T, Wilkes EH, Chakravarty P, Thompson R, Topping J, Cutillas PR, Lockley M, Serody JS, Capasso M, Balkwill FR. A strong B-cell response is part of the immune landscape in human high-grade serous ovarian metastases. Clin Cancer Res. 2017;23:250–262. doi: 10.1158/1078-0432.CCR-16-0081. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 84.de Visser KE, Korets LV, Coussens LM. De novo carcinogenesis promoted by chronic inflammation is B lymphocyte dependent. Cancer Cell. 2005;7:411–423. doi: 10.1016/j.ccr.2005.04.014. [DOI] [PubMed] [Google Scholar]
  • 85.Schioppa T, Moore R, Thompson RG, Rosser EC, Kulbe H, Nedospasov S, Mauri C, Coussens LM, Balkwill FR. B regulatory cells and the tumor-promoting actions of TNF-alpha during squamous carcinogenesis. Proc Natl Acad Sci U S A. 2011;108:10662–10667. doi: 10.1073/pnas.1100994108. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 86.Ammirante M, Luo JL, Grivennikov S, Nedospasov S, Karin M. B-cell-derived lymphotoxin promotes castration-resistant prostate cancer. Nature. 2010;464:302–5. doi: 10.1038/nature08782. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 87.Dieu-Nosjean MC, Antoine M, Danel C, Heudes D, Wislez M, Poulot V, Rabbe N, Laurans L, Tartour E, de Chaisemartin L, Lebecque S, Fridman WH, Cadranel J. Long-term survival for patients with non-small-cell lung cancer with intratumoral lymphoid structures. J. Clin. Oncol. 2008;26:4410–4417. doi: 10.1200/JCO.2007.15.0284. [DOI] [PubMed] [Google Scholar]
  • 88.Bindea G, Mlecnik B, Tosolini M, Kirilovsky A, Waldner M, Obenauf AC, Angell H, Fredriksen T, Lafontaine L, Berger A, Bruneval P, Fridman WH, Becker C, Pagès F, Speicher MR, Trajanoski Z, Galon J. Spatiotemporal dynamics of intratumoral immune cells reveal the immune landscape in human cancer. Immunity. 2013;39:782–795. doi: 10.1016/j.immuni.2013.10.003. [DOI] [PubMed] [Google Scholar]
  • 89.Germain C, Gnjatic S, Tamzalit F, Knockaert S, Remark R, Goc J, Lepelley A, Becht E, Katsahian S, Bizouard G, Validire P, Damotte D, Alifano M, Magdeleinat P, Cremer I, Teillaud JL, Fridman WH, Sautès-Fridman C, Dieu-Nosjean MC. Presence of B cells in tertiary lymphoid structures is associated with a protective immunity in patients with lung cancer. Am J Respir Crit Care Med. 2014;189:832–844. doi: 10.1164/rccm.201309-1611OC. [DOI] [PubMed] [Google Scholar]
  • 90.Nielsen JS, Sahota RA, Milne K, Kost SE, Nesslinger NJ, Watson PH, Nelson BH. CD20+ tumor-infiltrating lymphocytes have an atypical CD27- memory phenotype and together with CD8+ T cells promote favorable prognosis in ovarian cancer. Clin Cancer Res. 2012;18:3281–3292. doi: 10.1158/1078-0432.CCR-12-0234. [DOI] [PubMed] [Google Scholar]
  • 91.Kroeger DR, Milne K, Nelson BH. Tumor-infiltrating plasma cells are associated with tertiary lymphoid structures, cytolytic T-cell responses, and superior prognosis in ovarian cancer. Clin Cancer Res. 2016;22:3005–3015. doi: 10.1158/1078-0432.CCR-15-2762. [DOI] [PubMed] [Google Scholar]
  • 92.Goc J, Germain C, Vo-Bourgais TK, Lupo A, Klein C, Knockaert S, de Chaisemartin L, Ouakrim H, Becht E, Alifano M, Validire P, Remark R, Hammond SA, Cremer I, Damotte D, Fridman WH, Sautès-Fridman C, Dieu-Nosjean MC. Dendritic cells in tumor-associated tertiary lymphoid structures signal a Th1 cytotoxic immune contexture and license the positive prognostic value of infiltrating CD8+ T cells. Cancer Res. 2014;74:705–715. doi: 10.1158/0008-5472.CAN-13-1342. [DOI] [PubMed] [Google Scholar]
  • 93.Shah S, Divekar AA, Hilchey SP, Cho HM, Newman CL, Shin SU, Nechustan H, Challita-Eid PM, Segal BM, Yi KH, Rosenblatt JD. Increased rejection of primary tumors in mice lacking B cells: inhibition of anti-tumor CTL and TH1 cytokine responses by B cells. Int J Cancer. 2005;117:574–586. doi: 10.1002/ijc.21177. [DOI] [PubMed] [Google Scholar]
  • 94.Barbera-Guillem E, Nelson MB, Barr B, Nyhus JK, May KF Jr, Feng L, Sampsel JW. B lymphocyte pathology in human colorectal cancer. Experimental and clinical therapeutic effects of partial B cell depletion. Cancer Immunol Immunother. 2000;48:541–549. doi: 10.1007/PL00006672. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 95.Qin Z, Richter G, Schüler T, Ibe S, Cao X, Blankenstein T. B cells inhibit induction of T cell-dependent tumor immunity. Nat Med. 1998;4:627–630. doi: 10.1038/nm0598-627. [DOI] [PubMed] [Google Scholar]
  • 96.Yang C, Lee H, Jove V, Deng J, Zhang W, Liu X, Forman S, Dellinger TH, Wakabayashi M, Yu H, Pal S. Prognostic significance of B-cells and pSTAT3 in patients with ovarian cancer. PLoS One. 2013;8:e54029. doi: 10.1371/journal.pone.0054029. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 97.Biswas SK, Mantovani A. Macrophage plasticity and interaction with lymphocyte subsets: cancer as a paradigm. Nat Immunol. 2010;11:889–96. doi: 10.1038/ni.1937. [DOI] [PubMed] [Google Scholar]
  • 98.Mantovani A, Marchesi F, Porta C, Sica A, Allavena P. Inflammation and cancer: breast cancer as a prototype. Breast. 2007;16(Suppl 2):S27–33. doi: 10.1016/j.breast.2007.07.013. [DOI] [PubMed] [Google Scholar]
  • 99.Rubio C, Munera-Maravilla E, Lodewijk I, Suarez-Cabrera C, Karaivanova V, Ruiz-Palomares R, Paramio JM, Dueñas M. Macrophage polarization as a novel weapon in conditioning tumor microenvironment for bladder cancer: can we turn demons into gods? Clin Transl Oncol. 2019;21:391–403. doi: 10.1007/s12094-018-1952-y. [DOI] [PubMed] [Google Scholar]
  • 100.Zhu H, Leiss L, Yang N, Rygh CB, Mitra SS, Cheshier SH, Weissman IL, Huang B, Miletic H, Bjerkvig R, Enger PØ, Li X, Wang J. Surgical debulking promotes recruitment of macrophages and triggers glioblastoma phagocytosis in combination with CD47 blocking immunotherapy. Oncotarget. 2017;8:12145–12157. doi: 10.18632/oncotarget.14553. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 101.Sica A, Schioppa T, Mantovani A, Allavena P. Tumour-associated macrophages are a distinct M2 polarised population promoting tumour progression: potential targets of anti-cancer therapy. Eur J Cancer. 2006;42:717–727. doi: 10.1016/j.ejca.2006.01.003. [DOI] [PubMed] [Google Scholar]
  • 102.Mantovani A, Schioppa T, Porta C, Allavena P, Sica A. Role of tumor-associated macrophages in tumor progression and invasion. Cancer Metastasis Rev. 2006;25:315–322. doi: 10.1007/s10555-006-9001-7. [DOI] [PubMed] [Google Scholar]
  • 103.Allavena P, Sica A, Garlanda C, Mantovani A. The Yin-Yang of tumor-associated macrophages in neoplastic progression and immune surveillance. Immunol Rev. 2008;222:155–161. doi: 10.1111/j.1600-065X.2008.00607.x. [DOI] [PubMed] [Google Scholar]
  • 104.Kurahara H, Shinchi H, Mataki Y, Maemura K, Noma H, Kubo F, Sakoda M, Ueno S, Natsugoe S, Takao S. Significance of M2-polarized tumor-associated macrophage in pancreatic cancer. J Surg Res. 2011;167:e211–219. doi: 10.1016/j.jss.2009.05.026. [DOI] [PubMed] [Google Scholar]
  • 105.Chanmee T, Ontong P, Konno K, Itano N. Tumor-associated macrophages as major players in the tumor microenvironment. Cancers (Basel) 2014;6:1670–1690. doi: 10.3390/cancers6031670. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 106.Wan T, Liu JH, Zheng LM, Cai MY, Ding T. Prognostic significance of tumor-associated macrophage infiltration in advanced epithelial ovarian carcinoma. Ai Zheng. 2009;28:323–327. [PubMed] [Google Scholar]
  • 107.van Netten JP, George EJ, Ashmead BJ, Fletcher C, Thornton IG, Coy P. Macrophage-tumour cell associations in breast cancer. Lancet. 1993;342:872–873. [PubMed] [Google Scholar]
  • 108.Takanami I, Takeuchi K, Kodaira S. Tumor-associated macrophage infiltration in pulmonary adenocarcinoma: association with angiogenesis and poor prognosis. Oncology. 1999;57:138–142. doi: 10.1159/000012021. [DOI] [PubMed] [Google Scholar]
  • 109.Lan C, Huang X, Lin S, Huang H, Cai Q, Wan T, Lu J, Liu J. Expression of M2-polarized macrophages is associated with poor prognosis for advanced epithelial ovarian cancer. Technol Cancer Res Treat. 2013;12:259–267. doi: 10.7785/tcrt.2012.500312. [DOI] [PubMed] [Google Scholar]
  • 110.Cornelissen R, Lievense LA, Maat AP, Hendriks RW, Hoogsteden HC, Bogers AJ, Hegmans JP, Aerts JG. Ratio of intratumoral macrophage phenotypes is a prognostic factor in epithelioid malignant pleural mesothelioma. PLoS One. 2014;9:e106742. doi: 10.1371/journal.pone.0106742. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 111.Dzionek A, Fuchs A, Schmidt P, Cremer S, Zysk M, Miltenyi S, Buck DW, Schmitz J. BDCA-2, BDCA-3, and BDCA-4: three markers for distinct subsets of dendritic cells in human peripheral blood. J Immunol. 2000;165:6037–6046. doi: 10.4049/jimmunol.165.11.6037. [DOI] [PubMed] [Google Scholar]
  • 112.Liu YJ. IPC: professional type 1 interferon-producing cells and plasmacytoid dendritic cell precursors. Annu Rev Immunol. 2005;23:275–306. doi: 10.1146/annurev.immunol.23.021704.115633. [DOI] [PubMed] [Google Scholar]
  • 113.Kamath AT, Sheasby CE, Tough DF. Dendritic cells and NK cells stimulate bystander T cell activation in response to TLR agonists through secretion of IFN-alpha beta and IFN-gamma. J Immunol. 2005;174:767–776. doi: 10.4049/jimmunol.174.2.767. [DOI] [PubMed] [Google Scholar]
  • 114.Jego G, Palucka AK, Blanck JP, Chalouni C, Pascual V, Banchereau J. Plasmacytoid dendritic cells induce plasma cell differentiation through type I interferon and interleukin 6. Immunity. 2003;19:225–234. doi: 10.1016/s1074-7613(03)00208-5. [DOI] [PubMed] [Google Scholar]
  • 115.Zou W, Machelon V, Coulomb-L’Hermin A, Borvak J, Nome F, Isaeva T, Wei S, Krzysiek R, Durand-Gasselin I, Gordon A, Pustilnik T, Curiel DT, Galanaud P, Capron F, Emilie D, Curiel TJ. Stromal-derived factor-1 in human tumors recruits and alters the function of plasmacytoid precursor dendritic cells. Nat Med. 2001;7:1339–1346. doi: 10.1038/nm1201-1339. [DOI] [PubMed] [Google Scholar]
  • 116.Vermi W, Bonecchi R, Facchetti F, Bianchi D, Sozzani S, Festa S, Berenzi A, Cella M, Colonna M. Recruitment of immature plasmacytoid dendritic cells (plasmacytoid monocytes) and myeloid dendritic cells in primary cutaneous melanomas. J Pathol. 2003;200:255–268. doi: 10.1002/path.1344. [DOI] [PubMed] [Google Scholar]
  • 117.Wertel I, Polak G, Bednarek W, Barczyński B, Roliński J, Kotarski J. Dendritic cell subsets in the peritoneal fluid and peripheral blood of women suffering from ovarian cancer. Cytometry B Clin Cytom. 2008;74:251–258. doi: 10.1002/cyto.b.20410. [DOI] [PubMed] [Google Scholar]
  • 118.Curiel TJ, Cheng P, Mottram P, Alvarez X, Moons L, Evdemon-Hogan M, Wei S, Zou L, Kryczek I, Hoyle G, Lackner A, Carmeliet P, Zou W. Dendritic cell subsets differentially regulate angiogenesis in human ovarian cancer. Cancer Res. 2004;64:5535–5538. doi: 10.1158/0008-5472.CAN-04-1272. [DOI] [PubMed] [Google Scholar]
  • 119.Labidi-Galy SI, Sisirak V, Meeus P, Gobert M, Treilleux I, Bajard A, Combes JD, Faget J, Mithieux F, Cassignol A, Tredan O, Durand I, Ménétrier-Caux C, Caux C, Blay JY, Ray-Coquard I, Bendriss-Vermare N. Quantitative and functional alterations of plasmacytoid dendritic cells contribute to immune tolerance in ovarian cancer. Cancer Res. 2011;71:5423–5434. doi: 10.1158/0008-5472.CAN-11-0367. [DOI] [PubMed] [Google Scholar]
  • 120.Epstein LB, Shen JT, Abele JS, Reese CC. Sensitivity of human ovarian carcinoma cells to interferon and other antitumor agents as assessed by an in vitro semi-solid agar technique. Ann N Y Acad Sci. 1980;350:228–244. doi: 10.1111/j.1749-6632.1980.tb20624.x. [DOI] [PubMed] [Google Scholar]
  • 121.Einhorn N, Cantell K, Einhorn S, Strander H. Human leukocyte interferon therapy for advanced ovarian carcinoma. Am J Clin Oncol. 1982;5:167–172. doi: 10.1097/00000421-198204000-00064. [DOI] [PubMed] [Google Scholar]
  • 122.Biron CA. Interferons alpha and beta as immune regulators--a new look. Immunity. 2001;14:661–664. doi: 10.1016/s1074-7613(01)00154-6. [DOI] [PubMed] [Google Scholar]
  • 123.Wei S, Kryczek I, Zou L, Daniel B, Cheng P, Mottram P, Curiel T, Lange A, Zou W. Plasmacytoid dendritic cells induce CD8+ regulatory T cells in human ovarian carcinoma. Cancer Res. 2005;65:5020–5026. doi: 10.1158/0008-5472.CAN-04-4043. [DOI] [PubMed] [Google Scholar]
  • 124.Reizis B, Bunin A, Ghosh HS, Lewis KL, Sisirak V. Plasmacytoid dendritic cells: recent progress and open questions. Annu Rev Immunol. 2011;29:163–183. doi: 10.1146/annurev-immunol-031210-101345. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 125.Tjomsland V, Sandström P, Spångeus A, Messmer D, Emilsson J, Falkmer U, Falkmer S, Magnusson KE, Borch K, Larsson M. Pancreatic adenocarcinoma exerts systemic effects on the peripheral blood myeloid and plasmacytoid dendritic cells: an indicator of disease severity? BMC Cancer. 2010;10:87. doi: 10.1186/1471-2407-10-87. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 126.Li XF, Takiuchi H, Zou JP, Katagiri T, Yamamoto N, Nagata T, Ono S, Fujiwara H, Hamaoka T. Transforming growth factor-beta (TGF-beta)-mediated immunosuppression in the tumor-bearing state: enhanced production of TGF-beta and a progressive increase in TGF-beta susceptibility of anti-tumor CD4+ T cell function. Jpn J Cancer Res. 1993;84:315–325. doi: 10.1111/j.1349-7006.1993.tb02873.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 127.Jachimczak P, Bogdahn U, Schneider J, Behl C, Meixensberger J, Apfel R, Dörries R, Schlingensiepen KH, Brysch W. The effect of transforming growth factor-beta 2-specific phosphorothioate-anti-sense oligodeoxynucleotides in reversing cellular immunosuppression in malignant glioma. J Neurosurg. 1993;78:944–951. doi: 10.3171/jns.1993.78.6.0944. [DOI] [PubMed] [Google Scholar]
  • 128.Nabioullin R, Sone S, Mizuno K, Yano S, Nishioka Y, Haku T, Ogura T. Interleukin-10 is a potent inhibitor of tumor cytotoxicity by human monocytes and alveolar macrophages. J Leukoc Biol. 1994;55:437–442. doi: 10.1002/jlb.55.4.437. [DOI] [PubMed] [Google Scholar]
  • 129.Walker TM, Burger CJ, Elgert KD. Tumor growth alters T cell and macrophage production of and responsiveness to granulocyte-macrophage colony-stimulating factor: partial dysregulation through interleukin-10. Cell Immunol. 1994;154:342–357. doi: 10.1006/cimm.1994.1082. [DOI] [PubMed] [Google Scholar]
  • 130.Maeda H, Shiraishi A. TGF-beta contributes to the shift toward Th2-type responses through direct and IL-10-mediated pathways in tumor-bearing mice. J Immunol. 1996;156:73–78. [PubMed] [Google Scholar]
  • 131.Ren Y, Wu L, Frost AR, Grizzle W, Cao X, Wan M. Dual effects of TGF-beta on ERalpha-mediated estrogenic transcriptional activity in breast cancer. Mol Cancer. 2009;8:111. doi: 10.1186/1476-4598-8-111. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 132.Gold LI. The role for transforming growth factor-beta (TGF-beta) in human cancer. Crit Rev Oncog. 1999;10:303–360. [PubMed] [Google Scholar]
  • 133.Tomioka H, Sato K, Maw WW, Saito H. The role of tumor necrosis factor, interferon-gamma, transforming growth factor-beta, and nitric oxide in the expression of immunosuppressive functions of splenic macrophages induced by Mycobacterium avium complex infection. J Leukoc Biol. 1995;58:704–712. doi: 10.1002/jlb.58.6.704. [DOI] [PubMed] [Google Scholar]
  • 134.Zhang Q, Helfand BT, Carneiro BA, Qin W, Yang XJ, Lee C, Zhang W, Giles FJ, Cristofanilli M, Kuzel TM. Efficacy against human prostate cancer by prostate-specific membrane antigen-specific, transforming growth factor-beta insensitive genetically targeted CD8(+) T-cells derived from patients with metastatic castrate-resistant disease. Eur Urol. 2018;73:648–652. doi: 10.1016/j.eururo.2017.12.008. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 135.Sabat R. IL-10 family of cytokines. Cytokine Growth Factor Rev. 2010;21:315–324. doi: 10.1016/j.cytogfr.2010.11.001. [DOI] [PubMed] [Google Scholar]
  • 136.Zhou J, Ye F, Chen H, Lv W, Gan N. The expression of interleukin-10 in patients with primary ovarian epithelial carcinoma and in ovarian carcinoma cell lines. J Int Med Res. 2007;35:290–300. doi: 10.1177/147323000703500302. [DOI] [PubMed] [Google Scholar]
  • 137.Taylor A, Verhagen J, Blaser K, Akdis M, Akdis CA. Mechanisms of immune suppression by interleukin-10 and transforming growth factor-beta: the role of T regulatory cells. Immunology. 2006;117:433–442. doi: 10.1111/j.1365-2567.2006.02321.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 138.Mocellin S, Marincola FM, Young HA. Interleukin-10 and the immune response against cancer: a counterpoint. J Leukoc Biol. 2005;78:1043–1051. doi: 10.1189/jlb.0705358. [DOI] [PubMed] [Google Scholar]
  • 139.Napoletano C, Bellati F, Landi R, Pauselli S, Marchetti C, Visconti V, Sale P, Liberati M, Rughetti A, Frati L, Panici PB, Nuti M. Ovarian cancer cytoreduction induces changes in T cell population subsets reducing immunosuppression. J Cell Mol Med. 2010;14:2748–2759. doi: 10.1111/j.1582-4934.2009.00911.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 140.Mustea A, Könsgen D, Braicu EI, Pirvulescu C, Sun P, Sofroni D, Lichtenegger W, Sehouli J. Expression of IL-10 in patients with ovarian carcinoma. Anticancer Res. 2006;26:1715–1718. [PubMed] [Google Scholar]
  • 141.Jakowlew SB. Transforming growth factor-beta in cancer and metastasis. Cancer Metastasis Rev. 2006;25:435–457. doi: 10.1007/s10555-006-9006-2. [DOI] [PubMed] [Google Scholar]
  • 142.Muraoka-Cook RS, Dumont N, Arteaga CL. Dual role of transforming growth factor beta in mammary tumorigenesis and metastatic progression. Clin Cancer Res. 2005;11:937s–943s. [PubMed] [Google Scholar]
  • 143.Li X, Ye F, Chen H, Lu W, Wan X, Xie X. Human ovarian carcinoma cells generate CD4(+)CD25(+) regulatory T cells from peripheral CD4(+)CD25(-) T cells through secreting TGF-beta. Cancer Lett. 2007;253:144–153. doi: 10.1016/j.canlet.2007.01.024. [DOI] [PubMed] [Google Scholar]
  • 144.Itoh M, Takahashi T, Sakaguchi N, Kuniyasu Y, Shimizu J, Otsuka F, Sakaguchi S. Thymus and autoimmunity: production of CD25+CD4+ naturally anergic and suppressive T cells as a key function of the thymus in maintaining immunologic self-tolerance. J Immunol. 1999;162:5317–5326. [PubMed] [Google Scholar]
  • 145.Curotto de Lafaille MA, Lafaille JJ. CD4(+) regulatory T cells in autoimmunity and allergy. Curr Opin Immunol. 2002;14:771–778. doi: 10.1016/s0952-7915(02)00408-9. [DOI] [PubMed] [Google Scholar]
  • 146.Sakaguchi S, Sakaguchi N, Shimizu J, Yamazaki S, Sakihama T, Itoh M, Kuniyasu Y, Nomura T, Toda M, Takahashi T. Immunologic tolerance maintained by CD25+CD4+ regulatory T cells: their common role in controlling autoimmunity, tumor immunity, and transplantation tolerance. Immunol Rev. 2001;182:18–32. doi: 10.1034/j.1600-065x.2001.1820102.x. [DOI] [PubMed] [Google Scholar]
  • 147.Lavoué V, Thédrez A, Levêque J, Foucher F, Henno S, Jauffret V, Belaud-Rotureau MA, Catros V, Cabillic F. Immunity of human epithelial ovarian carcinoma: the paradigm of immune suppression in cancer. J Transl Med. 2013;11:147. doi: 10.1186/1479-5876-11-147. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 148.Shashikant L, Kesterson Joshua P. In pursuit of optimal cytoreduction in ovarian cancer patients: the role of surgery and surgeon. J Obstet Gynaecol India. 2009;59:209–216. [PMC free article] [PubMed] [Google Scholar]
  • 149.Fisher DT, Appenheimer MM, Evans SS. The two faces of IL-6 in the tumor microenvironment. Semin Immunol. 2014;26:38–47. doi: 10.1016/j.smim.2014.01.008. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 150.McLean K, Tan L, Bolland DE, Coffman LG, Peterson LF, Talpaz M, Neamati N, Buckanovich RJ. Leukemia inhibitory factor functions in parallel with interleukin-6 to promote ovarian cancer growth. Oncogene. 2019;38:1576–1584. doi: 10.1038/s41388-018-0523-6. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 151.West AJ, Tsui V, Stylli SS, Nguyen HPT, Morokoff AP, Kaye AH, Luwor RB. The role of interleukin-6-STAT3 signalling in glioblastoma. Oncol Lett. 2018;16:4095–4104. doi: 10.3892/ol.2018.9227. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 152.Culig Z, Puhr M. Interleukin-6 and prostate cancer: current developments and unsolved questions. Mol Cell Endocrinol. 2018;462:25–30. doi: 10.1016/j.mce.2017.03.012. [DOI] [PubMed] [Google Scholar]
  • 153.Tempfer C, Zeisler H, Sliutz G, Haeusler G, Hanzal E, Kainz C. Serum evaluation of interleukin 6 in ovarian cancer patients. Gynecol Oncol. 1997;66:27–30. doi: 10.1006/gyno.1997.4726. [DOI] [PubMed] [Google Scholar]
  • 154.Nowak M, Glowacka E, Szpakowski M, Szyllo K, Malinowski A, Kulig A, Tchorzewski H, Wilczynski J. Proinflammatory and immunosuppressive serum, ascites and cyst fluid cytokines in patients with early and advanced ovarian cancer and benign ovarian tumors. Neuro Endocrinol Lett. 2010;31:375–383. [PubMed] [Google Scholar]
  • 155.Kassim SK, El-Salahy EM, Fayed ST, Helal SA, Helal T, Azzam Eel-D, Khalifa A. Vascular endothelial growth factor and interleukin-8 are associated with poor prognosis in epithelial ovarian cancer patients. Clin Biochem. 2004;37:363–369. doi: 10.1016/j.clinbiochem.2004.01.014. [DOI] [PubMed] [Google Scholar]
  • 156.Keung EZ, Fairweather M, Raut CP. The role of surgery in metastatic gastrointestinal stromal tumors. Curr Treat Options Oncol. 2016;17:8. doi: 10.1007/s11864-015-0384-y. [DOI] [PubMed] [Google Scholar]
  • 157.Berger Y, Aycart S, Tabrizian P, Agmon Y, Mandeli J, Heskel M, Hiotis S, Sarpel U, Labow DM. Cytoreductive surgery and hyperthermic intraperitoneal chemotherapy in patients with liver involvement. J Surg Oncol. 2016;113:432–437. doi: 10.1002/jso.24153. [DOI] [PubMed] [Google Scholar]
  • 158.Cashin PH, Mahteme H, Spång N, Syk I, Frödin JE, Torkzad M, Glimelius B, Graf W. Cytoreductive surgery and intraperitoneal chemotherapy versus systemic chemotherapy for colorectal peritoneal metastases: a randomised trial. Eur J Cancer. 2016;53:155–162. doi: 10.1016/j.ejca.2015.09.017. [DOI] [PubMed] [Google Scholar]
  • 159.Huo YR, Richards A, Liauw W, Morris DL. Hyperthermic intraperitoneal chemotherapy (HIPEC) and cytoreductive surgery (CRS) in ovarian cancer: a systematic review and meta-analysis. Eur J Surg Oncol. 2015;41:1578–1589. doi: 10.1016/j.ejso.2015.08.172. [DOI] [PubMed] [Google Scholar]
  • 160.Chang SJ, Bristow RE, Chi DS, Cliby WA. Role of aggressive surgical cytoreduction in advanced ovarian cancer. J Gynecol Oncol. 2015;26:336–342. doi: 10.3802/jgo.2015.26.4.336. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 161.Noe A, Stewart GD, Bex A. The role of cytoreductive surgery in the era of targeted agents. Curr Opin Urol. 2015;25:374–380. doi: 10.1097/MOU.0000000000000192. [DOI] [PubMed] [Google Scholar]
  • 162.Bahra M, Pratschke J, Klein F, Neuhaus P, Boas-Knoop S, Puhl G, Denecke T, Pullankavumkal JR, Sinn M, Riess H, Pelzer U. Cytoreductive surgery for pancreatic cancer improves overall outcome of gemcitabine-based chemotherapy. Pancreas. 2015;44:930–936. doi: 10.1097/MPA.0000000000000365. [DOI] [PubMed] [Google Scholar]
  • 163.Motzer RJ, Mazumdar M, Bacik J, Berg W, Amsterdam A, Ferrara J. Survival and prognostic stratification of 670 patients with advanced renal cell carcinoma. J. Clin. Oncol. 1999;17:2530–2540. doi: 10.1200/JCO.1999.17.8.2530. [DOI] [PubMed] [Google Scholar]
  • 164.Motzer RJ, Hutson TE, Tomczak P, Michaelson MD, Bukowski RM, Rixe O, Oudard S, Negrier S, Szczylik C, Kim ST, Chen I, Bycott PW, Baum CM, Figlin RA. Sunitinib versus interferon alfa in metastatic renal-cell carcinoma. N Engl J Med. 2007;356:115–124. doi: 10.1056/NEJMoa065044. [DOI] [PubMed] [Google Scholar]
  • 165.Escudier B, Eisen T, Stadler WM, Szczylik C, Oudard S, Siebels M, Negrier S, Chevreau C, Solska E, Desai AA, Rolland F, Demkow T, Hutson TE, Gore M, Freeman S, Schwartz B, Shan M, Simantov R, Bukowski RM TARGET Study Group. Sorafenib in advanced clear-cell renal-cell carcinoma. N Engl J Med. 2007;356:125–134. doi: 10.1056/NEJMoa060655. [DOI] [PubMed] [Google Scholar]
  • 166.Sternberg CN, Davis ID, Mardiak J, Szczylik C, Lee E, Wagstaff J, Barrios CH, Salman P, Gladkov OA, Kavina A, Zarbá JJ, Chen M, McCann L, Pandite L, Roychowdhury DF, Hawkins RE. Pazopanib in locally advanced or metastatic renal cell carcinoma: results of a randomized phase III trial. J. Clin. Oncol. 2010;28:1061–1068. doi: 10.1200/JCO.2009.23.9764. [DOI] [PubMed] [Google Scholar]
  • 167.Hudes G, Carducci M, Tomczak P, Dutcher J, Figlin R, Kapoor A, Staroslawska E, Sosman J, McDermott D, Bodrogi I, Kovacevic Z, Lesovoy V, Schmidt-Wolf IG, Barbarash O, Gokmen E, O’Toole T, Lustgarten S, Moore L, Motzer RJ Global ARCC Trial. Temsirolimus, interferon alfa, or both for advanced renal-cell carcinoma. N Engl J Med. 2007;356:2271–2281. doi: 10.1056/NEJMoa066838. [DOI] [PubMed] [Google Scholar]

Articles from American Journal of Clinical and Experimental Urology are provided here courtesy of e-Century Publishing Corporation

RESOURCES