Loading

Medical Journal of Obstetrics and Gynecology

Aromatase Inhibitors or SERDs: Envisaging Roles in the Management of Cervical Cancer

Review Article | Open Access | Volume 8 | Issue 2

  • 1. Department of Microbiology, Kidwai Memorial Institute of Oncology, India
  • 2. Department of Gynecology, Kidwai Memorial Institute of Oncology, India
  • 3. Department of Radiation Oncology, Kidwai Memorial Institute of Oncology, India
+ Show More - Show Less
Corresponding Authors
Jayshree RS, Former Prof. and Head, Department of Microbiology, Kidwai Memorial Institute of Oncology, Hosur Road, Bangalore. 560029, B 906, Purva Heights, Bilekahalli, Bannerghatta Road, Bangalore, 560076. India, Tel: 9448059599.
Abstract

Considering that majority of patients with cervical cancer (CxCa) present with advanced disease, current five-year survival statistics of the disease is low; and chemo-radiation forms the mainstay of treatment. Immune tolerance as a hallmark of established tumors has come to stay; with a consequent renewal of interest in immunotherapy. Additionally, since >90% of CxCa is HPV driven, there is potential in immunotherapy targeting the viral oncoproteins. This could serve to enhance effector responses in the tumour microenvironment (TME), simultaneous to overcoming host immune suppression. Clinical response in oncology has dramatically improved following the use of immune check-point inhibitors in anticancer armamentarium. Considering that only a fraction of CxCa patients appear to benefit from the existing checkpoint agents, there is a need to further efforts towards characterizing newer immunotherapeutic targets in the TME. Contextually, there is substantial experimental, epidemiological and clinical evidence, implicating stromal estrogen in CxCa pathogenesis. Additionally, a lowered risk of developing cervical neoplasia was reported in women with carcinoma breast on anti-estrogen therapy. Also, in exvivo experiments, Selective Estrogen Receptor Disruptors (SERDs) were found to inhibit the function of tumor infiltrating regulatory T cells, Myeloid Derived Suppressor Cells and carcinoma associated fibroblasts derived from CxCa. Anti-estrogen therapy could therefore act as a checkpoint inhibitor in CxCa targetting locally produced estrogen in the TME. Here we review the potential immunomodulatory activity of aromatase inhibitors/SERDs which could pave the way for drugs interfering with estrogen signalling, some of which could be repurposed for better management of CxCa.

Citation

Jayshree RS, Adurthi S, Bafna UD, Mahesh Kumar M, Aradhana K (2020) Aromatase Inhibitors / SERDs: Envisaging Roles in the Management of Cervical Cancer. Med J Obstet Gynecol 8(2): 1136.

Keywords

•    Cancer cervix
•    Tumor stroma
•    Tumor microenvironment
•    Immune tolerance
•    Immune checkpoint inhibitors
•    Human Papilloma Virus
•    Selective Estrogen Receptor Disruptors (SERDs)
•    Regulatory T cells
•    Carcinoma-associated fibroblasts
•    Aromatase inhibitors
•    Immunotherapy
•    Myeloid-derived suppressor cells
•    Tumor-associated macrophages
•    Langerhans cells
•    Anti-CTLA4 antibodies
•    Anti PD1
•    Anti PDL1

ABBREVIATIONS

CxCa: Cancer Cervix; HPV: Human Papilloma Virus; SERDs: Selective Estrogen Receptor Disruptors; SERM: Selective Estrogen Receptor Modulator; TME: Tumor Microenvironment; Tregs: Regulatory T cells; CAFs: Carcinoma-associated Fibroblasts; AI: Aromatase Inhibitors; MDSCs: Myeloid-Derived Suppressor Cells; TAMs: Tumor- associated Macrophages; NK cells: Natural Killer Cells; CTLA-4; Cytotoxic T lymphocyte– Associated Antigen-4; PD-L1: Programmed Cell Death Ligand 1; ERα: Estrogen Receptor alpha; GPER: G Protein Coupled Receptor; PDE3A: Phosphodiesterase 3A; STS: Steroid Sulfatase; 17βHSD; 17 beta hydroxysteroid dehydrogenase; ICI: ICI 182,780; CTLs: Cytotoxic T lymphocytes; RU: RU 58668

INTRODUCTION-CERVICAL CANCER

In developing countries which lack both primary and secondary preventive programmes, cervical cancer (CxCa) continues to be a major cause of morbidity and mortality in women [1,2]. Poor prognosis of CxCa is primarily because majority of the cases present with advanced disease, which requires systemic treatment. Presently, the treatment for CxCa is concurrent platinum-based chemo-radiotherapy [3]. In patients with early stage CxCa (IB-IIB), chemo-radiation has been found to offer a 10-15% increase in survival at 5 years post-treatment compared to radiotherapy alone [4,5]. Survival statistics for the International Federation of Gynecology and Obstetrics (FIGO) stage I patients treated by surgery can be excellent: 5-year survival is 96, 95 and 80–93% in the UK, Germany and the USA, respectively [6]. However, more advanced cancers viz. FIGO stages II, III and IV, which are treated with platinum-based chemo-radiation, have lower 5-year survival rates [6]. Overall survival and prognosis of patients of stage IIIB CxCa in India is about 50% [7,8]. Also, current chemotherapy regimens offer response rates of only 35% to 50%, highlighting the need for bettering treatment strategies [9,10]. Hence there is a need for conceptualizing effective therapeutic vaccines against the disease [11], since this would help prevent about 5 million deaths that would occur in the next 20 years in women who are already infected with HPV [2].

THE TUMOR MICROENVIRONMENT

Solid tumour tissues can be divided into two distinct poorly-demarcated regions viz. the parenchyma (the tumour bed) and the stroma (the Tumor Microenvironment - TME). The tumor microenvironment is a hypoxic, acidic, and immune/inflammatory cell–enriched milieu that plays a crucial role in tumor development, growth, progression, and therapy resistance. The latter is a pathologically active niche that shapes tumor evolution and response to anticancer therapy including immunotherapy [12-16]. The TME encompasses a diverse spectrum of non-malignant cells viz. endothelial cells of blood and lymphatic vessels, mesenchymal stem cells, cancer-associated fibroblasts (CAFs), pericytes and infiltrating immune cells belonging to both the innate and adaptive arms of the immune system, along with the extracellular matrix and secreted soluble factors [12]. Exhaustive studies on the various components of the TME and their crosstalk with tumor cells have aided development of unique therapeutic strategies for improving outcomes in cancer. The vast landscape of intratumoral immune cells can broadly be divided into two groups. One is the protumorigenic / immunosuppressive populations of cells which play a major role in disrupting the capacity for the immune control of cancers: the major players being regulatory T cells (Tregs), Myeloid derived suppressor cells (MDSCs), the M2 polarized tumor associated macrophages (TAMs) etc. The other group comprising antitumorigenic/ effector cells capable of driving potent anti-tumour responses encompasses natural killer (NK) cells, dendritic cells and effector T cells (e.g. CD8+ Cytotoxic T lymphocytes - CTLs and CD4+Th1 cells, etc.). While both effectors and suppressors colocalize in the microenvironment, the immune scale in established cancers gets tilted towards immunoevasion [13]. Besides immunosuppressive cells, immunosuppressive co-inhibitory receptors like cytotoxic T lymphocyte–associated antigen-4 (CTLA-4), and programmed cell death ligand 1 (PD-L1); and immunosuppressive cytokines like IL10 and TGFβ are also active players in the battlefield [14].

IMMUNE INFILTRATES in CxCa

We had earlier reviewed the literature on immune infiltrates in cervical cancer [17]. Hence in this review, a brief discussion on immune infiltrates in CxCa would be restricted to cover published work only from June 2009 onwards, with an emphasis on Tregs. A large body of data demonstrates that CD4+ FOXP3+ Tregs are a strongly immunosuppressive subset of T cells which are a boon for the maintenance of immunological homeostasis and tolerance in infection, transplantation and autoimmunity but a bane for antitumor immunity [18-21]. Natural Tregs (nTregs) are phenotypically CD4+CD25hiCD127lo and express the lineage specific transcription factor FOXP3 - a master regulator which presides over the development, differentiation, maintenance and function of Tregs [22]. Increased frequencies of Treg cells in the TME appear to limit effector immune responses within the tumor, thereby preventing tumor control [23]. The TME of squamous cell carcinomas (SCC) of the cervix is marked by an immunoregulatory environment with increased expression of IDO1, immunosuppressive cytokines TGFβ and IL10; increased ratios of Kynurenine:Tryptophan; various types of Tregs – (both FOXP3+ and FOXP3 negative); reduced ratios of CD4+/ FOXP3+ cells and CD8+/FOXP3+ cells; low ratios of M1/M2 subsets of TAMs, Myeloid Derived Suppressor Cells (MDSCs) and anergic Langerhans cells [17,24-31]. Furthermore, such an immunosuppressive TME represented by high numbers of Tregs and PD-L1 expressing TAMs were found to permeate into metastatic lymph nodes (LNs), forming an immune-suppressive cordon around the tumour cells, enabling metastatic spread thereby resulted in a poor disease-free survival [32,33]. Additionally, exvivo experiments in our laboratory indicated that under the influence of the secretome of cervical CAFs (C-CAFs) naïve T cells differentiated into Tregs, while their differentiation into Th1, Th2 and Th17 subsets was inhibited (unpublished observation). Hence suppressing Treg cells in cancer immunotherapy is crucial for better prognosis. Additionally, considering the intrinsic antigenicity of CxCa, the prominence of Tregs as a therapeutic target is indisputable. Research studies have indicated that chemotherapeutic agents like cyclophosphamide exhibit anti-Treg properties and hence overall has an improved anticancer effect [34].

The advent of checkpoint inhibitors has heralded a rejuvenation of the field of anti-cancer immunotherapy since the latter boosts the ability of the immune system to recognize and destroy cancer cells, thus remarkably improving the prognosis of patients. Immune checkpoint inhibitor therapies targeting CTLA4 and PD-1 / PD-L1 enhance antitumor immunity by depleting CD4+ FOXP3+ Tregs and reducing their expansion in the TME. This may be one of the many other mechanisms that favour development of anti tumor immune responses [14,35-37]. One of the earliest targets used for decreasing intratumoral Tregs was CD25 – the IL2 receptor, a constitutive marker on many Treg subtypes [30]. In this model of CxCa, animals receiving anti-CD25 monoclonal antibody before receipt of the E7/Hsp70 DNA vaccine had higher numbers of E7 specific CD8+ T cells and more efficient control of tumors [30]. A resultant proliferation of CD8+ T cells, cytokine production and increase in the ratio of CD8+/Tregs has been recorded. These observations supported by results from several preclinical and early phase clinical studies formed the basis of Treg cell–targeted cancer immunotherapy [38,39]. Ipilimumab and tremelimumab, are two anti-CTLA4 antibodies, the former was the first to be approved by the FDA for the treatment of metastatic melanoma [40]. Likewise, Pembrolizumab and Nivolumab are monoclonal antibodies against PD-1 [41-43]. Currently Phase I/II clinical trials evaluating effects of anti-PD-1 therapy in CxCa are in progress [44-47]. Hence, based on the evidence that HPV positivity correlated with increased PD-L1 expression, checkpoint inhibitors are considered as a second line of treatment in cervical cancer [48]. Pembrolizumab has been approved by the FDA during or after chemotherapy for patients with recurrent or metastatic CxCa with progressive disease [48].

Other immune checkpoint targets under clinical trials are LAG-3, TIM-3 and CCR4 [44-45,49-52]. Antibodies to TIM 3 have an added advantage in anticancer therapy'

–  they specifically target tumor infiltrating Treg cells or tumor-associated dendritic cells with little action on extratumoral Tregs [42,43,47,49,53]. However, the response rates with existing check-point inhibitors which are majorly based on data from patients with metastatic disease [48], are low: only 20-30%; reflecting a need to undertake clinical studies on patients with early disease and explore novel targets for reversal of immunosuppression in the TME of CxCa.

ESTROGEN AND CANCER CERVIX

The uterine cervix is an estrogen (E2) responsive part of the female reproductive tract, the epithelium of which proliferates and differentiates physiologically under the influence of hormones during the menstrual cycle. Estradiol has been reported to use both the genomic and nongenomic pathways to signal a cell - by interacting with various receptors viz. Estrogen Receptor alpha (ERα), ERβ, membrane ERα, G protein coupled receptor (GPER), etc. In the canonical pathway, subsequent to binding to ERs, ligand- receptor complexes get translocated to the nucleus and ER interacts with estrogen responsive elements (ERE), on the DNA, following which epigenetic modifications and downstream gene activation ensues. The receptor thus functions as a transcription factor which regulates the expression of genes involved in cell survival and proliferation. The net outcome of these series of events is physiological alterations across various estrogen responsive tissues [54]. The non-genomic action is due to molecular crosstalk with growth factor pathways e.g. insulin growth factor, epidermal growth factor and fibroblast growth factor. Besides having a direct growth promoting action, in some tumors E2 is also pro-tumorigenic by virtue of its capability to modulate various cells of the TME [55].

HPV has long been recognized to be required but not sufficient to cause CxCa [56]. Of the various co-factors incriminated in cervical carcinogenesis E2 has been hypothesized to play a prominent role. Firstly, a series of pooled analysis of several case control studies have indicated that long-term use of exogenous estrogens in the form of hormonal contraceptives is associated with an increased risk of CxCa [57-61]. Likewise, several studies conducted in different continents indicated that exposure to endogenous estrogens by way of multiparity too increases the risk of squamous cell carcinoma of the cervix in HPV-infected women [57,60,62-63]. Various hypotheses explaining the observed relationship between estrogens, HPV and CxCa have been succinctly reviewed earlier [58]. While a marginal association between use of combined oral contraceptives (COC) and risk of acquisition of new HPV infections has been observed, evidence on COC likely promoting HPV persistence remain divided [58]. While physiological concentrations of E2 has been shown to induce expression of HPV oncogenes in cervical cancer cell lines [64-67], and promote the proliferation and inhibit apoptosis of cancer cells [58]; the phenomenon appears to be dose dependent – with high concentrations of the hormone as seen during pregnancy, stopping translation to promote apoptosis [68,69]. Secondly, both E2 and ERα were proven to be necessary to induce CxCa in the K14HPV16 transgenic mouse model of cervical carcinogenesis; and as a corollary Selective Estrogen Receptor Modulators (SERMs), were effective in controlling the development of cancer [70-74]. Thirdly, in an HPV18 transgenic mouse model, E2 was found to induce increased expression of HPV E6 and E7 oncogenes, - major driving force for cervical carcinogenesis [75]. Paradoxically though, in human disease, epithelial ERα expression, consistently declines with disease progression through cervical intraepithelial neoplasia (CIN) to invasive CxCa - both at the RNA and protein levels [76-82].

Earlier reviews incriminating E2 and ERα in cervical carcinogenesis assisted by HPV oncogenes have recommended the use of SERMs for treating the disease, arguing both for [83], and against the proposition [84]. The pro-apoptotic capability of non- physiological concentrations of E2 on CxCa cell line - HeLa was recently demonstrated [69]. Around the same time, using various receptor antagonists, another elegant study uncovered that in HeLa cells, E2 brought about apoptosis in a non-ER/non GPER fashion by interacting with phosphodiesterase 3A (PDE3A) - infact the latter has gained the distinction of being named as a new ER [68,85]. In this article, we review the role of E2/ERα in potentiating stromal cells and infiltrating immunosuppressive immune cells in cervical carcinogenesis. We earnestly hope that the article would provide food for thought for considering the use of antiestrogen therapy in the management of CxCa.

The stroma in the normal cervix has been shown to express ERα in as high as 93.7% [77], which is independent of changes in plasma hormonal concentrations seen during the menstrual cycle, perhaps indicating local synthesis [77,79,81,86]. Studies done in our laboratory showed that the microenvironment of CxCa was rich in the hormone E2, even in the absence of raised plasma levels (Figure 1A)- the distribution being mainly intracytoplasmic in the tumor epithelial cells and both intracytoplasmic and intranuclear in the stromal and infiltrating immune cells (Figure 1B) [87,88]. The enzyme aromatase too had a parallel distribution in the CxCa tissues thereby indicating that the hormone was being synthesized locally within the tumors (Figure 1B) [88,89]. In addition to aromatization of substrates, one needs to bear in mind that there are other alternative pathways of E2 synthesis in the tissues viz. from estrone sulfate (E1S), through Estrone (E1) by the actions of steroid sulfatase (STS), and 17 beta hydroxysteroid dehydrogenase (17β HSD) (Figure 2). This alternative pathway has been demonstrated in the cervical cancer cell line HeLa [90,91], although their role in the local synthesis of the hormone in clinical cases of CxCa still remains to be demonstrated.

About 30 to >50% of stromal cells express ERα in CIN and invasive squamous carcinomas of the uterine cervix. The distribution of the receptor in the stroma was uneven across the tumours, being independent of the stage of the disease and was surrounded by ERα-negative tumour cells [79,86-88,92]. Both fibroblasts and subsets of lymphocytes were amongst the cell types expressing the hormone receptor [87,88]. Raloxifene and fulvestrant (ICI 182,780 - ICI), are ER antagonists used in the treatment/prevention of human breast cancer and are classified under the category of SERMs and Selective Estrogen Receptor Disruptors (SERDs), respectively. Both the drugs efficiently cleared cancer and its precursor lesions in both cervix and vagina in K14E6/K14E7 double transgenic mice [83]. A subsequent posthoc analysis on the topic, however, ruled out the possibility of long term SERM treatment being useful in the prevention of Carcinoma In Situ (CIS) and CxCa in humans arguing that animal experiments could not be extrapolated to humans [84]. The objection may perhaps have been justified then, since in the mouse model but not in human CxCa, the tumor epithelium retained the expression of ERα. A recent review has discussed both the pro and anticancer effects of the steroid hormone in HPV positive cancers majorly focussing on the tumor epithelium, with minimal reference to the stroma and the immune infiltrates [93]. In an interesting observation, Ormeloxifine an oral contraceptive and a SERM have been advocated as a promising treatment for CxCa. The drug was demonstrated to block multiple signaling pathways particularly PI3k and Akt, inducing apoptosis of CxCa cell lines both in in vitro experiments and in an orthotopic animal model [94]. It would indeed be interesting to investigate the action of this SERM on tumor infiltrating immune cells considering that, this is also one of the pathways activated by E2 in Tregs amongst the many others [95].

The tumor epithelium however, cannot be viewed in isolation. A dynamic bidirectional cross talk between the three components viz. the HPV infected epithelium, the stroma and immune infiltrating cells plays a major role during carcinogenesis [71,96]. Drawing from their earlier work and using a sophisticated approach of stromal specific deletion of ERα, Lambert’s group proved that E2 signaling in the stromal cells drives tumorigenesis in the epithelium through paracrine mechanisms [82,97]. On similar lines, studies on gene expression profiling of exvivo cultured CxCa Associated Fibroblasts (C- CAFs), showed CAFs to express ERα and support tumor growth by promoting epithelial cell migration, proliferation, angiogenesis, metabolism, epithelial-to-mesenchymal transition and inflammation [87]. Further, using two categories of ER antagonists viz. a SERM and a SERD, modulation of genes associated with cell cycle and metabolism, affecting angiogenesis and cancer progression, was seen, proving thereby that the ERα signalling partly controlled the function of C-CAF [87]. HPV-dependent, E2-induced stromal genes are envisaged to be microenvironmental factors critical for cervical carcinogenesis [68,87]. Also, there is now growing evidence that molecular crosstalk between various components of the TME like CAFs and infiltrating immune cells can influence antitumor immunity [16]. Interestingly, as a consequence of all this research, stromal ERα singaling has been suggested as a therapeutic target for CxCa [98].

ESTROGEN IN INFILTRATING IMMUNE CELLS in CxCa

In addition to the direct carcinogenic action of E2 in CxCa, an indirect action of the sex steroid hormone, is induction of anti-inflammatory and regulatory immune responses which could potentiate HPV mediated cervical carcinogenesis [99]. Estradiol, is a powerful immunosuppressor and acts by: (i) recruiting MDSCs from the bone marrow into the spleen and tumor beds [100] (ii) augments the immunosuppressive activity of granulocytic MDSCs; via ERα [100] (iii) drives and polarizes infiltration of tumors with M2 type TAMs, (iv) promotes VEGF expression in them thus further enabling M2 recruitment; (v) increases the expression of granzyme B inhibitor - proteinase inhibitor-9 which counters the action of granzyme B mediated killing by NK cells and CTLs [55]. Pioneering studies in mice have proven that E2 administration expanded Tregs and induced overexpression of the transcription factor Foxp3 – a signature molecule of Tregs [101]. Additionally, a positive feedback loop between the hormone and infiltration of certain tumors by M2 type TAMs has been reported [55].

Cervical tumors harbour E2 [86-88], and Tregs (intra tumoral, draining LNs or circulating), had the highest intracellular levels of the hormone [88]. Estrogen is one of the various factors which regulate the functions of Tregs [102]. Probable mechanisms of action of estrogens could be modulation by signalling via ERα/ GPER [88,101,103-108]. Hence manipulation of E2 action using ER disruptors or modulators may unravel novel approaches to treat CxCa.

SERMS VS. SERDS IN THE MANAGEMENT CERVICAL CANCER

In ex-vivo experiments on tumor infiltrating and circulating Tregs, while SERDs (ICI and RU 58668), totally inhibited expression of both ERα and FOXP3 [88], MPP - a SERM, downregualted ERα, but failed to alter expression of FOXP3 (unpublished observation). Although the two SERDs – ICI and RU are chemically distinct, upon binding with ERα, the conformation of the receptor may be altered such that a similar protein – protein interaction surface is presented in both instances. Eventually, this may be flagging off the degradation of the ER-ligand complex by the proteasomal pathway [109]. Considering that (i). the conformation or shape of ER is determined by the chemical structure of the ligand which binds to it (ii) this binding directly affects the nuclear fate and protein turnover of ERα independently of its impact on transcription (iii) changes induced in the receptor upon binding to the ligand is ligand-specific (iv) this conformation is very crucial since it impacts protien – protein interactions and hence the ability of the receptor to interact with coregulatory proteins (i.e. coactivators and corepressors) (v). which eventually is critical to the regulation of target gene transcription (vi). locus specific variation in the engagement of coregulators has been observed for ERα. Hence it is quite likely that different ligands regulate engagement of specific ERα coregulators at the promoter of FOXP3 gene, which ultimately crucially affects the transcription of FOXP3 and hence the function of Tregs. Also, the relative expression of coactivators and corepressors, the subtype of ER, and its target gene promoter are known to affect the biocharacter of SERMs [110,111].

Therefore, considering the results of the effect of ICI on CAFs [87], Tregs [88], MDSCs [100], evidence of the efficacy of ICI in the treatment of CxCa in a mouse model [74], and our unpublished observation with SERM being ineffective in downregulating FOXP3, we opine that there is a strong case for undertaking clinical trials on the efficacy of SERDs in the management of CxCa. The crucial role played by the E2 pathway in the TME of various solid tumors has recently been highlighted [55]. Additional spinoffs of the use of ICI could be assisting the cytotoxic and chemo-radiosensitization actions of cisplatin [112,113].

SERDs (ICI and RU), majorly act through the genomic pathway of ER signaling in cancer cells to inhibit gene function [114]. We observed that intratumoral Treg cells also showed a direct influence of SERDs (ICI and RU) on the canonical ER signalling pathway - thus inhibiting FOXP3 expression and Treg cell function [88]. However, how and to what extent E2 influences the FOXP3 negative population of CxCa infiltrating Tregs still needs to be explored [25,115]. Additionally, ICI was also seen to inhibit the immunosuppressive function of granulocytic MDSCs in exvivo studies and orthotopic animal model of MDSCs in CxCa [100].

We also found ERα expression amongst infiltrating immune effector cells like CD4+ effectors, CD8+ CTLs in the TME although to a lower extent when compared to Tregs [88]. Hence while the action of ICI on other infiltrating immune cells also needs to be investigated, in all probability, limiting the availability of E2 in the TME would also help energize other subsets like Th1[103,116] and NK cells [117]. A positive flip side of the action of anti-estrogens is that they may also help regain granzyme B expression in HPV16-E7 expressing keratinocytes [118].

AROMATASE INHIBITORS IN THE MANAGEMENT OF CxCa

Arresting intratumoral E2 production with the use of aromatase inhibitors (AI), is a logical alternative to the use of SERDs, since in Tregs, E2 has also been shown to act non-genomically through membrane ER/GPER [95,106]. This was corroborated by our exvivo observation: E2 supplementation of ICI treated tumor infiltrating Tregs, partially reversed their suppressive function [88]. Considering that E2 influences Treg suppression by affecting multiple regulatory elements, both PD1 dependent, PD1 independent, FOXP3 dependent and FOXP3 independent pathways [101,103,104], blocking local production of E2 may be a more effective therapeutic option in the management of CxCa than blocking of ERs by way of using SERMs or SERDs. In support of the use of AI, is a recent population based study in breast cancer patients proving that long-term anti- estrogen use, including AI could reduce the incidence of cervical neoplasia [119]. Classically, protection offered against occurrence of high grade cervical dysplasia was seen only in women over the age of 50 years – which once again emphasizes local production as the main source of E2 in the tissues [91]. While Anastrozole has been shown to suppress the differentiation of naïve T cells into Tregs, increase IFNγ, IL12, and reduce IL4, IL10 in animals, Letrozole has been proven to be effective in reducing circulating Tregs in carcinoma breast patients responding to treatment [120,121]. While considering the use of AI, for targeting intratumoral synthesis of E2, one needs to consider other possible pathways of generation of E2 within the tumor tissues viz. from E1 - which is considered a major source of E2 in postmenopausal women [91]. This becomes relevant for designing therapeutics against the actions of 17−β HSD and STS (Figure 2) [122,123]. Also pertinent is the need to study common genetic polymorphism in CYP19A1 and ESR1 genes which would determine the variation in response to AIs and SERDs respectively

HPV THERAPEUTIC VACCINES

Cervical cancer is etiologically linked to persistent infection with high-risk HPV genotypes [124]. Therefore various immune based therapies including those targeting the viral oncogenes E6 and E7 have shown promising results in clinical trials in a spectrum of HPV-associated disease [11,125-128]. Agents that modulate the TME have an added advantage of being able to increase the efficacy of therapeutic vaccines [2]. There are a number of ways by which Tregs could be targeted viz.: by reducing their number, inhibiting their function, curtailing their influx into the TME and suppressing their generation in the periphery. Hence, we envisage that combining anti estrogen treatment with HPV therapeutic vaccines would be that magic bullet which would serve to be a three-pronged attack. Firstly, it would aid in reversing the immunosuppression in CxCa by inhibiting the suppressive function of intratumoral Tregs and MDSCs. Secondly, the function of CAFs in the tumor milieu would be simultaneously altered. Consequently, the specific immune response to the vaccines could be enhanced, thus promoting better immune control of tumors. Thirdly, AI use would perhaps check intratumoral E2 production which would perhaps also counter nongenomic signaling in cancer cells too.

We are prompted to classify antiestrogen therapy as a check-point inhibitor especially so, for CxCa. Whether it deserves the right to be called a universal check-point inhibitor for other tumors as well remains to be seen.

DISCUSSION & CONCLUSION

The steroid hormone E2 is a well-established pro-tumorigenic agent primarily by its direct genomic/non-genomic action on tumor cells. Estrogen has been considered a co- carcinogen, at least in the early stages of HPV mediated cervical carcinogenesis. Local production of E2 in CxCa tissues is partly through elaboration of aromatase. We have put forth this perspective on E2 being a crucial player in regulating the intratumoral immune response in CxCa by controlling the functions of CAFs, MDSCs and Tregs. Combining HPV oncoproteins with AI/SERD based immunotherapy and/or chemotherapy, on similar lines as of combination with chemotherapy/checkpoint inhibitors may be effective to counter Treg and MDSC mediated immunosuppression and help arrest growth of CAFs within tumors which would thereby improve the prognosis of CxCa [72,74,97,121,129-131]. Anti E2 treatment could thus become a new group of check-point blockade drugs functioning to antagonize intratumoral immunosuppression in CxCa. The need for clinical trials to evaluate the efficacy of AI and SERDs in the management of CxCa hence appears justified. As a prelude to clinical trials, comprehensive characterization of expression of ERα, aromatase, 17β-HSD, STS in the CxCa TME and their gene polymorphisms in large cohorts of patients across continents is warranted. In depth studies to understand the translational relevance of the proposed inhibitors is the need of the hour e.g. post treatment 19 evaluation of pharmacodynamic changes including those in the tumor immune infiltrate and associated clinical changes.

ACKNOWLEDGEMENTS

We wish to acknowledge the help of Drs. Mahua Sinha, Assistant Professor, Department of Microbiology, and Rekha V Kumar, Former Professor and Head, Histopathology and Medical Superintendent, Kidwai Memorial Institute of Oncology, Bangalore, India in editing the manuscript. This work was supported by grants from the Department of Biotechnology, Government of India (to RSJ) (no. BT/PR7014/ MED/14/927/2005), and the Indian Council of Medical Research (ICMR), India (No. 5/13/127/2011/NCD-III),; SA received a Senior Research fellowship from the Indian Council of Medical Research - (no. 3/2/2/72/2005/NCD-III); MMK was funded by the Council for Scientific and Industrial Research - Senior Research fellowship (no. 09/999 (0001)/2009-EMR-I).

REFERENCES

1. Bray F, Ferlay J, Soerjomataram I, Siegel RL, Torre LA, Jemal A. Global cancer statistics 2018: GLOBOCAN estimates of incidence and mortality worldwide for 36 cancers in 185 countries. CA Cancer J Clin. 2018; 68: 394-424.

2. Chabeda A, Yanez RJR, Lamprecht R, Meyers AE, Rybicki EP, Hitzeroth II. Therapeutic vaccines for high-risk HPV-associated diseases. Papillomavirus Res. 2018; 5: 46-58.

3. Takashi N, Tatsuya O, Hitoshi I, Yoshiyuki S, Takeo T. Current advancement in radiation therapy for uterine cervical cancer. J Radiat Res. 2010; 51: 1–8.

4. Eifel PJ, Winter K, Morris M, Levenback C, Grigsby PW, Cooper J, et al. Pelvic irradiation with concurrent chemotherapy versus pelvic and para-aortic irradiation for high-risk cervical cancer. J Clin Oncol. 2004; 22: 872-880.

5. Rose PG. Concurrent chemoradiation for locally advanced carcinoma of the cervix: where are we in 2006? Ann Oncol. 2006; 17 Suppl 10: x224-229.

6. Smola S, Trimble C, Stern PL. Human papillomavirus-driven immune deviation: challenge and novel opportunity for immunotherapy. Ther Adv Vaccines. 2017; 5: 69-82.

7. Swaminathan R, Rama R, Nalini S, Shanta V. Cancer survival in Chennai (Madras), India, 1990-1999. IARC Sci Publ. 2011; 162: 115-124.

8. Negi RR, Gupta M, Kumar M, Gupta MK, Seam R, Rastogi M. Concurrent chemoradiation in locally advanced carcinoma cervix patients. J Cancer Res Ther. 2010; 6: 159-166.

9. Ring KL, Pakish J, Jazaeri AA. Immune checkpoint inhibitors in the treatment of gynecologic malignancies. Cancer J. 2016; 22: 101-107.

10. Ryu HS. Concurrent chemoradiotherapy in cervical cancer (a new paradigm in cervical cancer treatment). Yonsei Med J. 2002; 43: 749- 753.

11. Ramanathan P, Dhandapani H, Jayakumar H, Seetharaman A, Thangarajan R. Immunotherapy for cervical cancer: Can it do another lung cancer? Curr Probl Cancer. 2018; 42: 148-160.

12. Hanahan D, Coussens LM. Accessories to the crime: Functions of cells recruited to the tumor microenvironment. Cancer Cell. 2012, 21, 309- 322.

13. Zitvogel L, Tesniere A, Kroemer G. Cancer despite immunosurveillance: immunoselection and immunosubversion. Nat Rev Immunol. 2006; 6: 715-727.

14. Shitara K, Nishikawa H. Regulatory T cells: a potential target in cancer immunotherapy. Ann N Y Acad Sci. 2018; 1417: 104-115.

15. 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.

16. Quail DF and Joyce JA. Microenvironmental regulation of tumor progression and metastasis. Nat. Med. 2013; 19: 1423-1437.

17. Jayshree RS, Sreenivas A, Tessy M, Krishna S. Cell intrinsic & extrinsic factors in cervical carcinogenesis. Indian J Med Res. 2009; 130: 286- 295.

18. Takeuchi Y, Nishikawa H. Roles of regulatory T cells in cancer immunity. Int Immunol. 2016; 8: 401-409.

19. Jang JE, Hajdu CH, Liot C, Miller G, Dustin ML, Bar-Sagi D. Crosstalk between Regulatory T Cells and Tumor-Associated Dendritic Cells Negates Anti-tumor Immunity in Pancreatic Cancer. Cell Rep. 2017; 20: 558-571.

20. Ward-Hartstonge KA, Kemp RA. Regulatory T-cell heterogeneity and the cancer immune response. Clin Transl Immunology. 2017; 6: e154.

21. Lee GR. Phenotypic and Functional Properties of Tumor-Infiltrating Regulatory T Cells. Mediators Inflamm. 2017; 2017: 5458178.

22. Ochs H D, Ziegler S F, Torgerson, TR. FOXP3 acts as a rheostat of the immune response. Immunol Rev. 2005; 203: 156-164.

23. Liu C, Workman CJ, Vignali DA. Targeting regulatory T cells in tumors. FEBS J. 2016; 283: 2731-2748.

24. Loddenkemper C, Hoffmann C, Stanke J, Nagorsen D, Baron U, Olek S, et al. Regulatory (FOXP3+) T cells as target for immune therapy of cervical intraepithelial neoplasia and cervical cancer. Cancer Sci. 2009; 100: 1112-1117.

25. Adurthi S, Geetashree Mukherjee, Krishnamurthy H, Krishna S, Bafna UD, Uma Devi K, Jayshree RS. Functional tumor infiltrating TH1 and TH2 effectors in large early-stage cervical cancer are suppressed by regulatory T cells. Int J Gynecol Cancer. 2012; 22: 1130-1137.

26. Kumar MM, Adurthi S, Ramachandran S, Mukherjee G, Joy O, Krishnamurthy H, et al. Toll-like receptors 7, 8, and 9 expression and function in primary human cervical cancer Langerhans cells: evidence of anergy. Int J Gynecol Cancer. 2013; 23: 184-192.

27. Heeren AM, de Boer E, Bleeker MC, Musters RJ, Buist MR, Kenter GG, et al. Nodal metastasis in cervical cancer occurs in clearly delineated fields of immune suppression in the pelvic lymph catchment area. Oncotarget. 2015; 6: 32484- 32493.

28. Hascitha J, Priya R, Jayavelu S, Dhandapani H, Selvaluxmy G, Sunder Singh S, et al. Analysis of Kynurenine/Tryptophan ratio and expression of IDO1 and 2 mRNA in tumour tissue of cervical cancer patients. Clin Biochem. 2016; 49: 919- 924.

29. Krishnan V, Schaar B, Tallapragada S, Dorigo O. Tumor associated macrophages in gynecologic cancers. Gynecol Oncol. 2018; 149: 205- 213.

30. Ao C, Zeng K. The role of regulatory T cells in pathogenesis and therapy of human papillomavirus-related diseases, especially in cancer. Infect Genet Evol. 2018; 65: 406-413.

31. Battaglia A, Buzzonetti A, Baranello C, Ferrandina G, Martinelli E, Fanfani F, et al. Metastatic tumour cells favour the generation of a tolerogenic milieu in tumour draining lymph node in patients with early cervical cancer. Cancer ImmunolImmunother. 2009; 58: 1363- 1373.

32. Heeren AM, Koster BD, Samuels S, Ferns DM, Chondronasiou D, Kenter GG, et al. High and interrelated rates of PD-L1+CD14+ antigen-presenting cells and regulatory T cells mark the microenvironment of metastatic lymph nodes from patients with cervical cancer. Cancer Immunol Res. 2015; 3: 48-58.

33. Heeren AM, Punt S, Bleeker MC, Gaarenstroom KN, van der Velden J, Kenter G G, et al. Prognostic effect of different PD-L1 expression patterns in squamous cell carcinoma and adenocarcinoma of the cervix. Mod Pathol. 2016; 29: 753-763.

34. Whiteside TL. The role of regulatory T cells in cancer immunology. Immunotargets Ther. 2015; 4: 159-171.

35. Selby MJ, Engelhardt JJ, Quigley M, Henning KA, Chen T, Srinivasan M, et al. Anti- CTLA-4 antibodies of IgG2a isotype enhance antitumor activity through reduction of intratumoral regulatory T cells. Cancer Immunol Res. 2013; 1: 32-42.

36. Simpson TR, Li F, Montalvo-Ortiz W, Sepulveda MA, Bergerhoff K, Arce F, et al. Fc- dependent depletion of tumor-infiltrating regulatory T cells co-defines the efficacy of anti -CTLA-4 therapy against melanoma. J Exp Med. 2013; 201: 1695-1710

37. DiDomenico J, Lamano JB, Oyon D, Li Y, Veliceasa D, Kaur G, et al. The immune checkpoint protein PD-L1 induces and maintains regulatory T cells in glioblastoma. Oncoimmunology. 2018; 7: e1448329.

38. Zou W. Regulatory T cells, tumour immunity and immunotherapy. Nat Rev Immunol. 2006; 6: 295-307.

39. Rice AE, Latchman YE, Balint JP, Lee JH, Gabitzsch ES, Jones, FR. An HPV- E6/E7 immunotherapy plus PD-1 checkpoint inhibition results in tumor regression and reduction in PD-L1 expression. Cancer Gene Ther. 2015; 22: 454-462.

40. De Felice F, Marchetti C, Palaia I, Ostuni R, Muzii L, Tombolini V, et al. Immune check-point in cervical cancer. Crit Rev Oncol Hematol. 2018; 129: 40-43.

41. Zappasodi R, Merghoub T, Wolchok JD. Emerging Concepts for Immune Checkpoint Blockade-Based Combination Therapies. Cancer Cell. 2018; 33: 581-598.

42. Zhang J, Burn C, Young K, Wilson M, Ly K, Budhwani M, et al. Microparticles produced by human papillomavirus type 16 E7- expressing cells impair antigen presenting cell function and the cytotoxic T cell response. Sci Rep. 2018; 8: 2373.

43. Zhang H, Li Y, Liu X, Liang Z, Yan M, Liu Q, et al. ImmTAC/Anti-PD-1 antibody combination to enhance killing of cancer cells by reversing regulatory T- cell-mediated immunosuppression. Immunology. 2018; 155: 238-250.

44. Kim JE., Patel MA, Mangraviti A, Kim ES, Theodros D, Velarde E, et al. Combination therapy with anti-PD-1, anti-TIM-3, and focal radiation results in regression of murine gliomas. Clin. Cancer Res. 2017; 23: 124-136.

45. Kim M, Kim H, Suh DH, Kim K, Kim H, Kim YB, et al. Identifying Rational Candidates for Immunotherapy Targeting PD-1/PD-L1 in Cervical Cancer. Anticancer Res. 2017; 37: 5087–5094.

46. Saglam O, Conejo-Garcia J. PD-1/PD-L1 immune checkpoint inhibitors in advanced cervical cancer. Integr Cancer Sci Ther. 2018; 5.

47. Dyer BA, Zamarin D, Eskandar RN, Mayadev JM. Role of Immunotherapy in the Management of Locally Advanced and Recurrent / Metastatic Cervical Cancer. J Natl Compr Canc Netw. 2019; 17: 91-97.

48. Liu Y, Wu L, Tong R, Yang F, Yin L, Li M, et al. PD-1/PD-L1 Inhibitors in Cervical Cancer. Front Pharmacol. 2019; 10: 65.

49. Sasidharan Nair V, Elkord E. Immune checkpoint inhibitors in cancer therapy: a focus on T-regulatory cells. Immunol Cell Biol. 2018; 96: 21-33.

50. Sabatos-Peyton CA, Nevin J, Brock A, Venable JD, Tan DJ, Kassam N, et al. Blockade of Tim-3 binding to phosphatidylserine and CEACAM1 is a shared feature of anti-Tim-3 antibodies that have functional efficacy. Oncoimmunology. 2017; 7: e1385690.

51. Shayan G, Srivastava R, Li J, Schmitt N, Kane LP, Ferris RL. Adaptive resistance to anti-PD1 therapy by Tim-3 upregulation is mediated by the PI3K-Akt pathway in head and neck cancer. Oncoimmunology. 2016; 6: e1261779.

52. Williams JB, Horton BL, Zheng Y, Duan Y, Powell JD, Gajewski TF. The EGR2 targets LAG-3 and 4-1BB describe and regulate dysfunctional antigen-specific CD8+ T cells in the tumor microenvironment. J Exp Med. 2017; 214: 381-400.

53. Liu JF, Wu L, Yang LL, Deng WW, Mao L, Wu H, et al. Blockade of TIM3 relieves immunosuppression through reducing regulatory T cells in head and neck cancer. J Exp Clin Cancer Res. 2018; 37: 44.

54. Baker JM, Al-Nakkash L, Herbst-Kralovetz MM. Estrogen-gut microbiome axis: Physiological and clinical implications. Maturitas. 2017; 103: 45-53.

55. Rothenberger NJ, Somasundaram A, Stabile LP. The Role of the Estrogen Pathway in the Tumor Microenvironment. Int J Mol Sci. 2018; 19: 611.

56. Munoz N, Castellsague X, de Gonzalez AB, Gissmann L. Chapter 1: HPV in the etiology of human cancer. Vaccine. 2006; 24 Suppl 3: S3-1-S310.

57. Muñoz N, Franceschi S, Bosetti C, Moreno V, Herrero R, Smith JS, et al. International Agency for Research on Cancer. Multicentric Cervical Cancer Study Group. Role of parity and human papillomavirus in cervical cancer: the IARC multicentric case-control study. Lancet. 2002; 359: 1093-101.

58. Marks M, Klein S, Gravitt P. Hormonal contraception and HPV: a tale of differing and overlapping mechanisms. Contraception. 2011; 2: 161- 174.

59. Marks MA, Gupta S, Liaw KL, Tadesse A, Kim E, Phongnarisorn C, et al. Prevalence and correlates of HPV among women attending family-planning clinics in Thailand. BMC Infect Dis. 2015; 15: 159.

60. Roura E, Travier N, Waterboer T, de Sanjosé S, Bosch FX, Pawlita M, et al. The Influence of Hormonal Factors on the Risk of Developing Cervical Cancer and Pre-Cancer: Results from the EPIC Cohort. PLoS One. 2016; 11: e0147029.

61. Jensen KE, Schmiedel S, Norrild B, Frederiksen K, Iftner T, Kjaer SK. Parity as a cofactor for high- grade cervical disease among women with persistent human papillomavirus infection: a 13-year follow- up. Br J Cancer. 2013; 108: 234-239.

62. Moreno V, Bosch FX, Muñoz N, Meijer CJ, Shah KV, Walboomers JM, et al. International Agency for Research on Cancer. Multicentric Cervical Cancer Study Group. Effect of oral contraceptives on risk of cervical cancer in women with human papillomavirus infection: the IARC multicentric case-control study. Lancet. 2002; 359: 1085-1092.

63. Rinaldi S, Plummer M, Biessy C, Castellsague X, Overvad K, Kruger KS, et al. Endogenous sex steroids and risk of cervical carcinoma: results from the EPIC study. Cancer Epidemiol Biomarkers Prev. 2011; 20: 2532-2540.

64. Chen YH, Huang LH, Chen TM. Differential effects of progestins and estrogens on long control regions of human papillomavirus types 16 and 18. Biochem Biophys Res Commun. 1996; 224: 651-659.

65. Mitrani-Rosenbaum S, Tsvieli R, Tur-Kaspa R. Oestrogen stimulates differential transcription of human papillomavirus type 16 in SiHa cervical carcinoma cells. J Gen Virol. 1989; 70: 2227-2232.

66. Kim CJ, Um SJ, Kim TY, Kim EJ, Park TC, Kim SJ, et al. Regulation of cell growth and HPV genes by exogenous estrogen in cervical cancer cells. Int J Gynecol Cancer. 2000; 10: 157-164.

67. Ruutu M, Wahlroos N, Syrjanen K, Johansson B, Syrjanen S. Effects of 17beta- estradiol and progesterone on transcription of human papillomavirus 16 E6/E7 oncogenes in CaSki and SiHa cell lines. Int J Gynecol Cancer. 2006; 16: 1261-1268.

68. Li D, Chen J, Ai Y, Gu X, Li L, Che D, et al. Estrogen-Related Hormones Induce Apoptosis by Stabilizing Schlafen-12 Protein Turnover. Mol. Cell. 2019; 75: 1103-1116.

69. Bristol ML, James CD, Wang X, Fontan CT, Morgan IM. Estrogen Attenuates the Growth of Human Papillomavirus-Positive Epithelial Cells. mSphere. 2020; 5: e00049-20.

70. Arbeit JM, Howley PM, Hanahan D. Chronic estrogen-induced cervical and vaginal squamous carcinogenesis in human papillomavirus type 16 transgenic mice. Proc Natl Acad Sci USA. 1996; 93: 2930-2935.

71. Spurgeon ME, Lambert PF. Human Papillomavirus and the Stroma: Bidirectional Crosstalk during the Virus Life Cycle and Carcinogenesis. Viruses. 2017; 9: E219.

72. Chung SH, Wiedmeyer K, Shai A, Korach KS, Lambert PF. Requirement for estrogen receptor alpha in a mouse model for human papillomavirus- associated cervical cancer. Cancer Res. 2008; 68: 9928-9934.

73. Riley RR, Duensing S, Brake T, Münger K, Lambert PF, Arbeit JM. Dissection of human papillomavirus E6 and E7 function in transgenic mouse models of cervical carcinogenesis. Cancer Res. 2003; 63: 4862- 4871.

74. Chung SH, Lambert PF. Prevention and treatment of cervical cancer in mice using estrogen receptor antagonists. Proc Natl Acad Sci USA. 2009; 106: 19467-19472.

75. Park JS, Rhyu JW, Kim CJ, Kim HS, Lee SY, Kwon YI, et al. Neoplastic change of squamo-columnar junction in uterine cervix and vaginal epithelium by exogenous estrogen in hpv-18 URR E6/E7 transgenic mice. Gynecol Oncol. 2003; 89: 360-368.

76. Zhai Y, Bommer GT, Feng Y, Wiese AB, Fearon ER, Cho KR. Loss of estrogen receptor 1 enhances cervical cancer invasion. Am J Pathol. 2010; 177: 884-1895.

77. López-Romero R, Garrido-Guerrero E, Rangel-López A, ManuelApolinar L, Piña-Sánchez P, Lazos-Ochoa M, et al. The cervical malignant cells display a down regulation of ER-α but retain the ER-β expression. Int J Clin Exp Pathol. 2013; 6: 1594-1602.

78. Nikolaou M, Koumoundourou D, Ravazoula P, Papadopoulou M, Michail G, Decavalas G. An immunohistochemical analysis of sexsteroid receptors, tumor suppressor gene p53 and Ki-67 in the normal and neoplastic uterine cervix squamous epithelium. Med Pregl. 2014; 67: 202-207.

79. Nonogaki H, Fujii S, Konishi I, Nanbu Y, Ozaki S, Ishikawa Y, et al. Estrogen receptor localization in normal and neoplastic epithelium of the uterine cervix. Cancer. 1990; 66: 2620-2627.

80. Konishi I, Fujii S, Nonogaki H, Nanbu Y, Iwai T, Mori T. Immunohistochemical analysis of estrogen receptors, progesterone receptors, Ki-67 antigen, and human papillomavirus DNA in normal and neoplastic epithelium of the uterine cervix. Cancer. 1991; 68: 1340-1350.

81. Coelho FR, Prado JC, Pereira Sobrinho JS, Hamada G, Landman G, Pinto CA, et al. Estrogen and progesterone receptors in human papilloma virus-related cervical neoplasia. Braz J Med Biol Res. 2004; 37: 83-88.

82. den Boon JA, Pyeon D, Wang SS, Horswill M, Schiffman M, Sherman M, et al. Molecular transitions from papillomavirus infection to cervical precancer and cancer: Role of stromal estrogen receptor signaling. Proc Natl Acad Sci USA. 2015; 112: E3255-E3264.

83. Chung SH, Franceschi S, Lambert PF. Estrogen and ER alpha: culprits in cervical cancer? Trends Endocrinol Metab. 2010; 21: 504-511.

84. Castle PE. Do selective estrogen receptor modulators treat cervical precancer and cancer? Time to pool data from relevant trials. Int J Cancer. 2011; 128: 997-998.

85. Lamb HM, Hardwick JM. The Dark Side of Estrogen Stops Translation to Induce Apoptosis. Mol Cell. 2019; 75: 1087-1089.

86. Mosny DS, Herholz J, Degen W, Bender HG. Immunohistochemical investigations of steroid receptors in normal and neoplastic squamous epithelium of the uterine cervix. Gynecol Oncol. 1989; 35: 373-377.

87. Kumar MM, Davuluri S, Poojar S, Mukherjee G, Bajpai AK, Bafna UD, et al. Role of estrogen receptor alpha in human cervical cancer-associated fibroblasts: a transcriptomic study. Tumour Biol. 2016; 37: 4409-4420.

88. Adurthi S, Kumar MM, Vinodkumar HS, Geetashree Mukherjee H. Krishnamurthy, Acharya Kshitish K, et al. Oestrogen Receptor-- binds the FOXP3 promoter and modulates regulatory T-cell function in human cervical cancer. Sci Rep. 2017; 7: 17289-17304.

89. Nair, HB Luthra R, Kirma N, Liu YG, Flowers L, Evans D, et al. Induction of aromatase expression in cervical carcinomas: effects of endogenous estrogen on cervical cancer cell proliferation. Cancer Res. 2005; 65: 11164-11173.

90. Fournier MA, Poirier D. Estrogen formation in endometrial and cervix cancer cell lines: involvement of aromatase, steroid sulfatase and 17beta-hydroxysteroid dehydrogenases (types 1, 5, 7 and 12). Mol Cell Endocrinol. 2009; 301: 142-145.

91. Gruber CJ, Tschugguel W, Schneeberger C, Huber JC. Production and actions of estrogens. N Engl J Med. 2002; 346: 340-352.

92. Kwasniewska A, Postawski K, Gozdzicka-Jozefiak A, Kwasniewski W, Grywalska E, Zdunek M, et al. Estrogen and progesterone receptor expression in HPV-positive and HPV-negative cervical carcinomas. Oncol Rep. 2011; 26: 53-160.

93. James CD, Morgan IM, Bristol ML. The Relationship between EstrogenRelated Signaling and Human Papillomavirus Positive Cancers. Pathogens. 2020; 9: 403.

94. Chauhan N, Maher DM, Yallapu MM, Bilal B Hafeez, Man M Singh, Subhash C, Chauhan, et al. A triphenylethylene nonsteroidal SERM attenuates cervical cancer growth. Sci Rep. 2019; 9: 10917.

95. Prieto GA, Rosenstein Y. Oestradiol potentiates the suppressive function of human CD4+CD25+ regulatory T cells by promoting their proliferation. Immunology. 2006; 118: 58-65.

96. De Nola R, Menga A, Castegna A, Vera Loizzi, Girolamo Ranieri, Ettore Cicinelli, et al. The Crowded Crosstalk between Cancer Cells and Stromal Microenvironment in Gynecological Malignancies: Biological Pathways and Therapeutic Implication. Int J Mol Sci. 2019; 20: 2401.

97. Chung SH, Shin MK, Korach KS, Lambert PF. Requirement for stromal estrogen receptor alpha in cervical neoplasia. Horm Cancer. 2013; 4: 50-59.

98. Son J, Park Y, Chung SH. Epithelial oestrogen receptor α is dispensable for the development of oestrogen-induced cervical neoplastic diseases. J Pathol. 2018; 245: 147-152.

99. Marks MA, Gravitt PE, Burk RD, Studentsov Y, Farzadegan H, Klein SL. Progesterone and 17 beta-estradiol enhance regulatory responses to human papillomavirus type 16 virus-like particles in peripheral blood mononuclear cells from healthy women. Clin Vaccine Immunol. 2010; 17: 609-617.

100. Kozasa K, Mabuchi S, Matsumoto Y, Hiromasa Kuroda, Eriko Yokoi, Naoko Komura, et al. Estrogen stimulates female cancer progression by inducing myeloid-derived suppressive cells: investigations on pregnant and non-pregnant experimental models. Oncotarget. 2019; 10: 1887-1902.

101. Polanczyk MJ, Carson BD, Subramanian S, Afentoulis M, Vandenbark AA, Ziegler SF, et al. Cutting edge: Estrogen drives expansion of the CD4+CD25+ regulatory T cell compartment. J Immunol. 2004; 173: 2227-2230.

102. Yang XF. Factors regulating apoptosis and homeostasis of CD4+ CD25 (high) FOXP3+ regulatory T cells are new therapeutic targets. Front Biosci. 2008; 13: 1472-1499.

103. Polanczyk M J, Hopke C, Vandenbark AA, Offner H. Estrogen-mediated immunomodulation involves reduced activation of effector T cells, potentiation of Treg cells, and enhanced expression of the PD-1 costimulatory pathway. J Neurosci Res. 2006; 84: 370-378

104. Polanczyk MJ, Hopke C, Vandenbark AA, Offner H. Treg suppressive activity involves estrogen dependent expression of programmed death-1 (PD-1). Int Immunol. 2007; 19: 337-343.

105. Tai P, Wang J, Jin H, Song X, Yan J, Kang Y, et al. Induction of regulatory T cells by physiological level estrogen. J Cell Physiol. 2008; 214: 456- 464.

106. Yates MA, Li Y, Chlebeck PJ, Offner H. GPR30, but not estrogen receptor-alpha, is crucial in the treatment of experimental autoimmune encephalomyelitis by oral ethinyl estradiol. BMC Immunol. 2010; 11: 20-24.

107. Luo CY, Wang L, Sun C, Li DJ. Estrogen enhances the functions of CD4(+)CD25(+)Foxp3(+) regulatory T cells that suppress osteoclast differentiation and bone resorption in vitro. Cell Mol Immunol. 2011; 8: 50-58.

108. Valor L, Teijeiro R, Aristimuño C, Faure F, Alonso B, de Andrés C, et al. Estradiol-dependent perforin expression by human regulatory T-cells. Eur J Clin Invest. 2011; 41: 357-364.

109. Bryan M. Wittmann, Andrea Sherk and Donald P. McDonnell. Definition of functionally important mechanistic differences among selective estrogen receptor down-regulators. Cancer Res. 2007; 67: 9549-9560.

110. Dutertre M, Smith CL. Molecular mechanisms of selective estrogen receptor modulator (SERM) action. J Pharmacol Exp Ther. 2000; 295: 431-437.

111. Kocanova Silvia, Mahta Mazaheri, Stéphanie Caze-Subra, Kerstin Bystricky. Ligands specify estrogen receptor alpha nuclear localization and degradation. BMC Cell Biol. 2010; 11: 98.

112. García-López P, Rodríguez-Dorantes M, Enrique Perez-Cardenas E, Cerbon M, Mohar-Betancourt A. Synergistic effects of ICI 182,780 on the cytotoxicity of cisplatin in cervical carcinoma cell lines. Cancer Chemother Pharmacol. 2004; 53: 533–540.

113. Segovia-Mendoza M, Jurado R, Mir R, Medina LA, Prado-Garcia H, Garcia-Lopez P. Antihormonal agents as a strategy to improve the effect of chemo-radiation in cervical cancer: in vitro and in vivo study. BMC Cancer. 2015; 15: 21.

114. Traboulsi T, El Ezzy M, Gleason JL, Mader S. Antiestrogens: structure-activity relationships and use in breast cancer treatment. J Mol Endocrinol. 2017; 58: R15-R31.

115. Filaci G, Fenoglio D, Fravega M, Ansaldo G, Borgonovo G, Traverso P, et al. CD8+CD28- T regulatory lymphocytes inhibiting T cell proliferative and cytotoxic functions infiltrate human cancers. J Immunol. 2007; 179: 4323-4334.

116. Polese B, Gridelet V, Araklioti E, Martens H, Perrier d’Hauterive S, Geenen V. The Endocrine Milieu and CD4 T Lymphocyte Polarization during Pregnancy. Front Endocrinol (Lausanne). 2014; 5: 106-117.

117. Jiang X, Ellison SJ, Alarid ET, Shapiro DJ. Interplay between the levels of estrogen and estrogen receptor controls the level of the granzyme inhibitor, proteinase inhibitor 9 and susceptibility to immune surveillance by natural killer cells. Oncogene. 2007; 26: 4106-4114.

118. Munguía-Moreno JA, Díaz-Chavéz J, García-Villa E, Albino-Sanchez ME, Mendoza-Villanueva D, Ocadiz-Delgado R, et al. Early synergistic interactions between the HPV16-E7 oncoprotein and 17β-oestradiol for repressing the expression of Granzyme B in a cervical cancer model. Int J Oncol. 2018; 53: 579-591.

119. Hsieh CJ, Hong MK, Chen PC, Wang JH, Chu TY. Antiestrogen use reduces risk of cervical neoplsia in breast cancer patients: a population-based study. Oncotarget. 2017; 8: 29361-29369.

120. Ray A, Ficek M. Immunomodulatory effects of anti-estrogenic drugs. Acta Pharm. 2012; 62: 141-155.

121. Generali D, Bates G, Berruti A, Brizzi MP, Campo L, Bonardi S, et al. Immunomodulation of FOXP3+ regulatory T cells by the aromatase inhibitor letrozole in breast cancer patients. Clin Cancer Res. 2009; 15: 1046-1051.

122. Purohit A, Foster PA. Steroid sulfatase inhibitors for estrogen- and androgen-dependent cancers. J Endocrinol. 2012; 212: 99-110.

123. Secky L, Svoboda M, Klameth L, Bajna E, Hamilton G, Zeillinger R, et al. The sulfatase pathway for estrogen formation: targets for the treatment and diagnosis of hormone-associated tumors. J Drug Deliv. 2013; 2013: 957605.

124. Dillner J. Prevention of human papillomavirus-associated cancers. Semin Oncol. 2015; 42: 272-283.

125. Maldonado L, Teague JE, Morrow MP, Jotova I, Wu TC, Wang C, et al. Intramuscular therapeutic vaccination targeting HPV16 induces T cell responses that localize in mucosal lesions. Sci Transl Med. 2014; 6: 221ra13.

126. Trimble CL, Morrow MP, Kraynyak KA, Shen X, Dallas M, Yan J, et al. Safety, efficacy, and immunogenicity of VGX-3100, a therapeutic synthetic DNA vaccine targeting human papillomavirus 16 and 18 E6 and E7 proteins for cervical intraepithelial neoplasia 2/3: a randomised, double-blind, placebo-controlled phase 2b trial. Lancet. 2015; 386: 2078-2088.

127. Stevanovi? S, Draper LM, Langhan MM, Campbell TE, Kwong ML, Wunderlich JR, et al. Complete regression of metastatic cervical cancer after treatment with human papilloma virus targeted tumor-infiltrating T cells. J Clin Oncol. 2015; 33: 1543-1550.

128. Draper LM, Kwong ML, Gros A, Stevanovi? S, Tran E, Kerkar S, et al. Targeting of HPV-16+ epithelial cancer cells by TCR gene engineered T cells directed against E6. Clin Cancer Res. 2015; 21: 4431-4439.

129. Spurgeon ME, den Boon JA, Horswill M, Sonalee Barthakur, Omid Forouzan, Janet S Rader, et al. Human papillomavirus oncogenes reprogram the cervical cancer microenvironment independently of and synergistically with estrogen. Proc Natl Acad Sci USA. 2017; 114: E9076-E9085.

130. Weir GM, Hrytsenko O, Stanford MM, Berinstein NL, Karkada M, Liwski RS, et al. Metronomic cyclophosphamide enhances HPV16E7 peptide vaccine induced antigen-specific and cytotoxic T-cell mediated antitumor immune response. Oncoimmunology. 2014; 3: e953407.

131. Zhao J, Zeng W, Cao Y, Liang X, Huang B. Immunotherapy of HPV infection- caused genital warts using low dose cyclophosphamide. Expert Rev Clin Immunol. 2014; 10: 791-799.

 

Jayshree RS, Adurthi S, Bafna UD, Mahesh Kumar M, Aradhana K (2020) Aromatase Inhibitors / SERDs: Envisaging Roles in the Management of Cervical Cancer. Med J Obstet Gynecol 8(2): 1136

Received : 20 Jul 2020
Accepted : 14 Aug 2020
Published : 17 Aug 2020
Journals
Annals of Otolaryngology and Rhinology
ISSN : 2379-948X
Launched : 2014
JSM Schizophrenia
Launched : 2016
Journal of Nausea
Launched : 2020
JSM Internal Medicine
Launched : 2016
JSM Hepatitis
Launched : 2016
JSM Oro Facial Surgeries
ISSN : 2578-3211
Launched : 2016
Journal of Human Nutrition and Food Science
ISSN : 2333-6706
Launched : 2013
JSM Regenerative Medicine and Bioengineering
ISSN : 2379-0490
Launched : 2013
JSM Spine
ISSN : 2578-3181
Launched : 2016
Archives of Palliative Care
ISSN : 2573-1165
Launched : 2016
JSM Nutritional Disorders
ISSN : 2578-3203
Launched : 2017
Annals of Neurodegenerative Disorders
ISSN : 2476-2032
Launched : 2016
Journal of Fever
ISSN : 2641-7782
Launched : 2017
JSM Bone Marrow Research
ISSN : 2578-3351
Launched : 2016
JSM Mathematics and Statistics
ISSN : 2578-3173
Launched : 2014
Journal of Autoimmunity and Research
ISSN : 2573-1173
Launched : 2014
JSM Arthritis
ISSN : 2475-9155
Launched : 2016
JSM Head and Neck Cancer-Cases and Reviews
ISSN : 2573-1610
Launched : 2016
JSM General Surgery Cases and Images
ISSN : 2573-1564
Launched : 2016
JSM Anatomy and Physiology
ISSN : 2573-1262
Launched : 2016
JSM Dental Surgery
ISSN : 2573-1548
Launched : 2016
Annals of Emergency Surgery
ISSN : 2573-1017
Launched : 2016
Annals of Mens Health and Wellness
ISSN : 2641-7707
Launched : 2017
Journal of Preventive Medicine and Health Care
ISSN : 2576-0084
Launched : 2018
Journal of Chronic Diseases and Management
ISSN : 2573-1300
Launched : 2016
Annals of Vaccines and Immunization
ISSN : 2378-9379
Launched : 2014
JSM Heart Surgery Cases and Images
ISSN : 2578-3157
Launched : 2016
Annals of Reproductive Medicine and Treatment
ISSN : 2573-1092
Launched : 2016
JSM Brain Science
ISSN : 2573-1289
Launched : 2016
JSM Biomarkers
ISSN : 2578-3815
Launched : 2014
JSM Biology
ISSN : 2475-9392
Launched : 2016
Archives of Stem Cell and Research
ISSN : 2578-3580
Launched : 2014
Annals of Clinical and Medical Microbiology
ISSN : 2578-3629
Launched : 2014
JSM Pediatric Surgery
ISSN : 2578-3149
Launched : 2017
Journal of Memory Disorder and Rehabilitation
ISSN : 2578-319X
Launched : 2016
JSM Tropical Medicine and Research
ISSN : 2578-3165
Launched : 2016
JSM Head and Face Medicine
ISSN : 2578-3793
Launched : 2016
JSM Cardiothoracic Surgery
ISSN : 2573-1297
Launched : 2016
JSM Bone and Joint Diseases
ISSN : 2578-3351
Launched : 2017
JSM Bioavailability and Bioequivalence
ISSN : 2641-7812
Launched : 2017
JSM Atherosclerosis
ISSN : 2573-1270
Launched : 2016
Journal of Genitourinary Disorders
ISSN : 2641-7790
Launched : 2017
Journal of Fractures and Sprains
ISSN : 2578-3831
Launched : 2016
Journal of Autism and Epilepsy
ISSN : 2641-7774
Launched : 2016
Annals of Marine Biology and Research
ISSN : 2573-105X
Launched : 2014
JSM Health Education & Primary Health Care
ISSN : 2578-3777
Launched : 2016
JSM Communication Disorders
ISSN : 2578-3807
Launched : 2016
Annals of Musculoskeletal Disorders
ISSN : 2578-3599
Launched : 2016
Annals of Virology and Research
ISSN : 2573-1122
Launched : 2014
JSM Renal Medicine
ISSN : 2573-1637
Launched : 2016
Journal of Muscle Health
ISSN : 2578-3823
Launched : 2016
JSM Genetics and Genomics
ISSN : 2334-1823
Launched : 2013
JSM Anxiety and Depression
ISSN : 2475-9139
Launched : 2016
Clinical Journal of Heart Diseases
ISSN : 2641-7766
Launched : 2016
Annals of Medicinal Chemistry and Research
ISSN : 2378-9336
Launched : 2014
JSM Pain and Management
ISSN : 2578-3378
Launched : 2016
JSM Women's Health
ISSN : 2578-3696
Launched : 2016
Clinical Research in HIV or AIDS
ISSN : 2374-0094
Launched : 2013
Journal of Endocrinology, Diabetes and Obesity
ISSN : 2333-6692
Launched : 2013
Journal of Substance Abuse and Alcoholism
ISSN : 2373-9363
Launched : 2013
JSM Neurosurgery and Spine
ISSN : 2373-9479
Launched : 2013
Journal of Liver and Clinical Research
ISSN : 2379-0830
Launched : 2014
Journal of Drug Design and Research
ISSN : 2379-089X
Launched : 2014
JSM Clinical Oncology and Research
ISSN : 2373-938X
Launched : 2013
JSM Bioinformatics, Genomics and Proteomics
ISSN : 2576-1102
Launched : 2014
JSM Chemistry
ISSN : 2334-1831
Launched : 2013
Journal of Trauma and Care
ISSN : 2573-1246
Launched : 2014
JSM Surgical Oncology and Research
ISSN : 2578-3688
Launched : 2016
Annals of Food Processing and Preservation
ISSN : 2573-1033
Launched : 2016
Journal of Radiology and Radiation Therapy
ISSN : 2333-7095
Launched : 2013
JSM Physical Medicine and Rehabilitation
ISSN : 2578-3572
Launched : 2016
Annals of Clinical Pathology
ISSN : 2373-9282
Launched : 2013
Annals of Cardiovascular Diseases
ISSN : 2641-7731
Launched : 2016
Journal of Behavior
ISSN : 2576-0076
Launched : 2016
Annals of Clinical and Experimental Metabolism
ISSN : 2572-2492
Launched : 2016
Clinical Research in Infectious Diseases
ISSN : 2379-0636
Launched : 2013
JSM Microbiology
ISSN : 2333-6455
Launched : 2013
Journal of Urology and Research
ISSN : 2379-951X
Launched : 2014
Journal of Family Medicine and Community Health
ISSN : 2379-0547
Launched : 2013
Annals of Pregnancy and Care
ISSN : 2578-336X
Launched : 2017
JSM Cell and Developmental Biology
ISSN : 2379-061X
Launched : 2013
Annals of Aquaculture and Research
ISSN : 2379-0881
Launched : 2014
Clinical Research in Pulmonology
ISSN : 2333-6625
Launched : 2013
Journal of Immunology and Clinical Research
ISSN : 2333-6714
Launched : 2013
Annals of Forensic Research and Analysis
ISSN : 2378-9476
Launched : 2014
JSM Biochemistry and Molecular Biology
ISSN : 2333-7109
Launched : 2013
Annals of Breast Cancer Research
ISSN : 2641-7685
Launched : 2016
Annals of Gerontology and Geriatric Research
ISSN : 2378-9409
Launched : 2014
Journal of Sleep Medicine and Disorders
ISSN : 2379-0822
Launched : 2014
JSM Burns and Trauma
ISSN : 2475-9406
Launched : 2016
Chemical Engineering and Process Techniques
ISSN : 2333-6633
Launched : 2013
Annals of Clinical Cytology and Pathology
ISSN : 2475-9430
Launched : 2014
JSM Allergy and Asthma
ISSN : 2573-1254
Launched : 2016
Journal of Neurological Disorders and Stroke
ISSN : 2334-2307
Launched : 2013
Annals of Sports Medicine and Research
ISSN : 2379-0571
Launched : 2014
JSM Sexual Medicine
ISSN : 2578-3718
Launched : 2016
Annals of Vascular Medicine and Research
ISSN : 2378-9344
Launched : 2014
JSM Biotechnology and Biomedical Engineering
ISSN : 2333-7117
Launched : 2013
Journal of Hematology and Transfusion
ISSN : 2333-6684
Launched : 2013
JSM Environmental Science and Ecology
ISSN : 2333-7141
Launched : 2013
Journal of Cardiology and Clinical Research
ISSN : 2333-6676
Launched : 2013
JSM Nanotechnology and Nanomedicine
ISSN : 2334-1815
Launched : 2013
Journal of Ear, Nose and Throat Disorders
ISSN : 2475-9473
Launched : 2016
JSM Ophthalmology
ISSN : 2333-6447
Launched : 2013
Journal of Pharmacology and Clinical Toxicology
ISSN : 2333-7079
Launched : 2013
Annals of Psychiatry and Mental Health
ISSN : 2374-0124
Launched : 2013
Annals of Pediatrics and Child Health
ISSN : 2373-9312
Launched : 2013
JSM Clinical Pharmaceutics
ISSN : 2379-9498
Launched : 2014
JSM Foot and Ankle
ISSN : 2475-9112
Launched : 2016
JSM Alzheimer's Disease and Related Dementia
ISSN : 2378-9565
Launched : 2014
Journal of Addiction Medicine and Therapy
ISSN : 2333-665X
Launched : 2013
Journal of Veterinary Medicine and Research
ISSN : 2378-931X
Launched : 2013
Annals of Public Health and Research
ISSN : 2378-9328
Launched : 2014
Annals of Orthopedics and Rheumatology
ISSN : 2373-9290
Launched : 2013
Journal of Clinical Nephrology and Research
ISSN : 2379-0652
Launched : 2014
Annals of Community Medicine and Practice
ISSN : 2475-9465
Launched : 2014
Annals of Biometrics and Biostatistics
ISSN : 2374-0116
Launched : 2013
JSM Clinical Case Reports
ISSN : 2373-9819
Launched : 2013
Journal of Cancer Biology and Research
ISSN : 2373-9436
Launched : 2013
Journal of Surgery and Transplantation Science
ISSN : 2379-0911
Launched : 2013
Journal of Dermatology and Clinical Research
ISSN : 2373-9371
Launched : 2013
JSM Gastroenterology and Hepatology
ISSN : 2373-9487
Launched : 2013
Annals of Nursing and Practice
ISSN : 2379-9501
Launched : 2014
JSM Dentistry
ISSN : 2333-7133
Launched : 2013
Author Information X