Skip to main content

Research advances on the immune research and prospect of immunotherapy in pituitary adenomas

Abstract

Background

Pituitary adenomas are one type of intracranial tumor, which can be divided into microadenoma (≤ 1 cm), macroadenoma (> 1 cm), and giant adenoma (≥ 4 cm) according to their diametral sizes. They are benign, typically slow-progressing, whereas the biological behavior of some of them is invasive, which presents a major clinical challenge. Treatment of some pituitary adenomas is still difficult due to drug resistance or multiple relapses, usually after surgery, medication, and radiation. At present, no clear prediction and treatment biomarkers have been found in pituitary adenomas and some of them do not cause clinical symptoms, so patients are often found to be ill through physical examination, and some are even found through autopsy. With the development of research on pituitary adenomas, the immune response has become a hot spot and may serve as a novel disease marker and therapeutic target.

The distribution and function of immune cells and their secreted molecules in pituitary adenomas are extremely complex. Researchers found that infiltration of immune cells may have a positive effect on the treatment and prognosis of pituitary adenomas. In this review, we summarized the advance of tumor immunity in pituitary adenomas, revealing the immunity molecules as potential biomarkers as well as therapeutic agents for pituitary adenomas.

Conclusion

The immune studies related to pituitary adenomas may help us find relevant immune markers. At the same time, the exploration of immunotherapy also provides new options for the treatment of pituitary adenomas.

Introduction

Pituitary adenomas (PAs) are benign intracranial tumors with the third highest incidence, accounting for about 10–15% of intracranial tumors [1]. However, up to 20% of PAs appear clinically invasive symptoms, showing rapid growth and recurrence after treatment [2, 3]. More seriously, studies have shown that 0.2% of PAs are more likely to become pituitary cancer [4]. PAs can lead to clinical symptoms by oppressing the normal pituitary gland and invading the cavernous sinus or skull base structure. In addition, a variety of “functioning” PAs secrete supraphysiologic levels of hormones, resulting in profound systemic effects that reflect the changes in hormone levels [5]. At present, the treatment of PAs mainly depends on surgical resection [6]. However, the complete resection rate of tumors is only 66 to 78% [7]. When surgery and chemotherapy fail, radiotherapy becomes the treatment of choice against PAs. But radiation strikes at healthy tissue. For example, it can lead to visual impairment, hypopituitary, and cerebrospinal fluid leakage. Therefore, this clinical dilemma has inspired researchers to find new markers and treatment methods [8, 9]. With the deepening understanding of the tumor environment and its development, immunotherapy is a promising alternative therapy for the treatment of drug-resistant or recurrent PAs [9]. The mechanism of PAs occurrence and development is still unclear, which may be a result of multiple factors such as epigenetics, genes, and tumor microenvironment(TME) [10, 11]. The TME is a special environment generated by the interaction between tumor cells and the host during tumor development [12]. It is a complex environment consisting of fibroblasts, myofibroblasts, endothelial cells, immune cells, and extracellular matrix (ECM) which can affect tumor proliferation, invasiveness, and angiogenesis [13, 14]. Current research has focused on immune cells in TME. A tumor is a systemic disease in which inflammatory immune cells, chemokines, and cytokines influence tumor growth and invasion [15]. The infiltrating immune cells in brain tissue include macrophages, neutrophils, T cells, natural killer cells(NK cells), and other immune cells. Recent studies have shown that they play an important role in brain function and physiology; they can influence behavior and participate in the pathogenesis of various neuropathologies [16]. Myeloid cells, such as tumor-associated macrophages, dendritic cells, and lymphocytes, such as T cells and B cells, make up the tumor microenvironmental immune cells (TMICs) (Fig. 1) [17]. They may be located in the core, margin, or adjacent tertiary lymphoid structures (TLS) of the tumor [18]. The role of these infiltrating immune cells and their secreted molecules is complex and can ultimately lead to tumor-promoting or anti-tumor effects through interaction with the tumor and its host [19, 20]. There are still few studies on the immunity of pituitary adenoma. In this review, we summarize the current immunological studies on pituitary adenoma to point out the prospect that immunity molecules may be helpful to prognosis prediction and clinical targeted therapy.

Fig. 1
figure 1

Immune cells infiltrate in pituitary adenoma. Immune cells infiltrate in PAs. The role of immune cells in PAs invasion, migration, and proliferation is unclear

Immune cells infiltrate in PAs

Innate immunity (non-specific immunity) and adaptive immunity(specific immunity)comprise the human immune system, and adaptive immunity involves T and B cells and their secreted factors [21]. As part of the natural immune cell population, NK cells can control tumor growth by interacting with other immune cells and tumor cells [22, 23]. For example, MHC-I offer CD8+T lymphocytes cell antigens, such as from its protein and virus protein antigen, and extracellular antigen by MHC II usually provided to CD4+T lymphocyte, loss of MHC-I during the process of tumor formation is one typical method that cancer cells evade monitoring of CD8+T cells, NK cells express to identify MHC-I molecules-inhibition of cell surface receptors, and eliminate cannot fully express a large number of MHC-I molecular targets [24,25,26]. Current studies have shown that although the number of NK cells in solid tumors is smaller than that of CD8+T cells, CD4+T cells, and B cells, the presence of NK cells in TME may be associated with a good prognosis [27, 28]. Macrophages can be polarized into M1 or M2 macrophages. Tumor-associated macrophages(TAMs) are generally characterized as M2-like macrophages [29]. By stimulating tumor angiogenesis and inhibiting the anti-tumor immune response mediated by T cells, TAMs promote the proliferation, invasion, and metastasis of tumor cells [30]. Recent studies by Zhang et al. have shown that PA may produce excess lactate, resulting in TME acidification, which reshapes TAMs into an M2-type phenotype and then secrets CCL17 through TAMs to enhance tumor invasion through the CCL17/CCR4/mTORC1 axis [31]. Meanwhile, M2 macrophages may play a role in neoangiogenesis in pituitary adenomas together with B cells, CD4 +T cells, and Foxp3 + lymphocytes [32]. Dendritic cells (DCs) are the main antigen-presenting cells and serve as a bridge between adaptive and innate immune systems [33]. DCs promotes tumorigenesis in some tumors and inhibits tumorigenesis in others, which indicates the phenomenon of tumor stage dependence, that is, DCs show tumor inhibition effect in the early stage, but with the tumor promotion, it can be changed to promote the development of the tumor [33]. A key part of the adaptive immune response is CD4+T and CD8+T cells [34, 35]. Current studies have shown that CD4+T cells mediate anti-tumor responses through a variety of mechanisms, such as the CCR5 ligand that acts as the center of CD4+ and CD8+T cell activation. CD8+T cells destroy target cells by differentiating into cytotoxic T cells (CTLS), releasing cytotoxic particles [36]. In addition, B cell subsets have immunosuppressive and/or regulatory functions and may play a key role in regulating the human immune response to tumors [37].

Research conducted by Wang et al. showed that there is a different distribution of tumor-infiltration immune cells (TIICs) between PAs and the normal pituitary gland, and there are also differences among different PA subtypes. In addition, three immune clusters can be identified among PAs according to their distribution, and each one of PAs shows unique characteristics [38]. PAs can be classified into functional pituitary adenomas (FPAs) and non-functional pituitary adenomas (NFPAs) according to the hormone secretion [39]. FPAs include prolactinomas, somatotroph, corticotroph, thyrotrophin, and rarely gonadotroph adenomas [40]. Existing studies have shown that immune cells infiltrate differently in both FPAs and NFPAs [14, 41,42,43]. According to imaging characteristics and pathological diagnosis, PAs can be divided into invasive PAs and non-invasive PAs [44, 45]. There are also inconsistencies in the distribution of infiltrating immune cells in PAs, depending on the aggressiveness [46, 47]. For example, the infiltration of CD8+T cells was positively correlated with PRL and PRL and GH immunostaining in the tumor and was also positively correlated with the invasions of PAs [41, 48]. The research of recent years has been recently reviewed by us and is presented in Table 1 [5, 14, 32, 41, 42, 47,48,49,50,51,52,53,54,55,56,57,58,59]. It is worth noting that the role of immune cells in PAs is not yet clear and may vary among different tumor stages and types. Therefore, before the exact role of immune cells in pituitary tumors is understood, it is necessary to define the immune characteristics of PAs [59].

Table 1 Immune studies of PAs

Inflammatory factors associated with PAs

Current research has shown that cytokines including interferon (IFN), interleukin insurance-linked securities (ILs), and tumor necrosis factor (TNF) play a key role in the differentiation of the pituitary gland and oncogenesis of PAs [60, 61]. T helper type 1 (Th1) cells secrete TNF-α, IFN-γ, and IL-2, and they induce cell-mediated immune responses, whereas Th2 cells secrete IL-4, IL-5, IL-6, IL-10, and IL-13 [60]. IFN is divided into type I and type II. The main classes of type I IFN are IFN-α, IFN-β, IFN-ɛ, IFN-κ, and IFN-ω. IFN-γ belongs to type II IFN [62]. IFNs play a regulatory role in pituitary hormone secretion. Both stimulatory and inhibitory effects of IFNs (IFNα and IFNγ) on the secretion of ACTH, PRL, and GH have been reported [63]. IFN-α significantly inhibits hormone secretion and intracellular hormone concentration in human GH secreting PAs, prolactin, and NFPAs or gonadotropin adenoma [63]. TNF can trigger a variety of potential outcomes through TNFR1 and TNFR2 activation signals, including cell proliferation, gene activation, or cell death [64]. In invasive PAs, TNF-α promotes pathological osteoclast formation by directly inducing osteoclast differentiation, leading to inflammatory bone destruction [65]. Therefore, Zhu et al. proposed that TNF-α might be a novel target in the treatment of osteo-invasive pituitary adenoma [66]. TNF-α also may be an important regulator of hemorrhagic transformation in pituitary adenomas indicated by the finding that hemorrhagic pituitary adenomas displayed higher protein and mRNA levels of TNF-α [67]. The function of ILs is related to the expression and regulation of the immune response, which is involved in many factors originating from lymphocytes or macrophages. Qiu et al. reported that in the serum of patients with PAs (include invasive and non-invasive PAs), IL-4, IL-5, and IL-17A were significantly increased, while Th1/Th2 ratio was significantly decreased [60]. Notably, serum IL-17A levels in patients with invasive PA reported in this study were significantly higher than those in patients with non-invasive PA. While another study showed no significant difference between serum IL-17A levels and PA growth types [68]. In addition, the serum level of IL-4 was significantly higher in patients with idiopathic hyperprolactinemia [69]. The bidirectional role of IL-6 in PAs has also been reported. Paracrine IL-6 may be a condition that permits the growth of pituitary cells by contributes to excessive hormone production, growth, and neovascularization of pituitary adenomas, while autocrine IL-6 inhibits the aggressive growth and malignant transformation of tumors [70]. In vitro studies, an earlier study showed that IL-2 and IL-6 stimulated the proliferation of GH3 cells [71]. Furthermore, other inflammatory factors present in TME also play complex roles that require further detailed exploration by researchers. For example, macrophage migration inhibitory factor (MIF) is an immunomodulator that can be induced by pituitary hormones, enhancing the production of inflammatory cytokines such as TNF, IL-1, and IFN, to play its role of anti-tumor or tumor promotion [72].

Status of immunotherapy for PAs

The relationship between tumor cells and the immune system is divided into three stages according to the theory proposed by Jun et al., at the initial stage, tumor cells are recognized and cleared by immune cells. As tumors develop, there is a phase of balance between tumor cells and immune cells. Eventually, the immune response is evaded by the tumor cells, and the immune system is unable to cope with the tumors [73]. As the tumor grows and changes, tumor cells can evade the immune system, leading to further spread, infiltration, and even metastasis. This kind of avoidance is achieved by the co-stimulation and co-inhibition signals of tumor cells [74]. The immune checkpoint is a regulator of immune activation and plays an important role in maintaining self-tolerance, controlling immune response intensity, and reducing tissue damage [75]. Immune checkpoints can be used by tumors to suppress T cell activation. The most representative ones are the co-inhibitory cytotoxic T lymphocyte-associated protein 4 (CTLA-4) and programmed cell death 1 (PD-1) pathways. T cell dysfunction, failure, and neutralization in tumors can be caused by activation of the PD-1 signaling pathway [76]. CTLA-4 is expressed in various types of tumors including PAs and can play a role by limiting the CD4+T cell phenotype [77, 78]. As a result, blocking immune checkpoints is a new approach to the treatment of many types of tumors [79]. PD-L1 binds to the PD-1 receptor on activated T cells and inhibits the cytotoxic anti-tumor function of T cells while blocking this interaction can produce a lasting T cell response [80,81,82]. Blocking CTLA-4 receptors on lymphocytes leads to T cell activation, which reduces tumor-mediated immune tolerance [83]. When the CTLA-4 and PD-1 are blocked, the stimulation signal of T cells are activated, the number of cytotoxic T cells with anti-tumor activity increase, the production and proliferation of pro-inflammatory cytokines can also be promoted, and finally, the tumor destruction can be accelerated [84, 85]. While anti-programmed cell death protein 1 (anti-PD-1) and anti-cytotoxic T-lymphocyte-associated protein 4 (anti-CTLA4) antibodies have been extensively used to target immune checkpoints in many cancers, their use in pituitary tumors has just commenced.

Recently, it has been reported that PD-L1 expression is higher in pituitary tumors invading the cavernous sinus [86]. In addition, the expression of PD-L1 also significantly increased with the increase of serum GH, PRL, ACTH, and cortisol levels [42]. Similar conditions also exist in the expression of PD-1 [49, 52]. However, no significant difference was found in CTLA4 expression [52]. In animal experiments, Hanna R et al. provided a new theoretical basis for the immunotherapy of PAs, that is, anti-PD-L1 treatment successfully reduced the plasma ACTH level of model mice, reduced the growth of PAs, and improved the survival rate of model mice [5]. In clinical application, a patient with ACTH-secreting pituitary carcinoma who received clinical remission using ipilimumab (anti-CTLA-4) and nivolumab (anti-PD-1) ICI was reported by Sol et al. [87]. Moreover, immune checkpoints such as TIM3 and LAG3 are also potential targets for immunotherapy of pituitary adenomas [88]. In addition to immune checkpoints, other methods of immunotherapy are being explored. Hazrati et al. published a report of a female with a macroprolactinoma refractory to conventional therapy and was successfully treated with immunotherapy. A Th1 activator adjuvant was inoculated with autoantigens weekly for 24 weeks, and following this therapy, her serum prolactin levels decreased and adenoma almost disappeared [89]. However, the side effects of immunotherapy are varied and any organ (skin, intestines, liver, and glands like the thyroid and adrenal glands) may be affected, leading to a variety of diseases (rash, pruritus, vitiligo, diarrhea, colitis, hepatitis, hypophysitis, hypothyroidism, primary adrenal insufficiency, and diabetes) [90, 91]. So, further studies are needed to refine the immunotherapy regimen for PAs. For example, immunotherapy combined with radiation therapy may be a promising option [92].

Future perspectives and conclusions

At present, the specific mechanism of the interaction between pituitary adenoma and the human immune system is not clear, and studies are limited to the apparent expression level of related markers. However, based on the existing evidence, namely, the expression of immune cells and the changes and differences of inflammatory factors in pituitary adenomas, the immune research on pituitary adenomas deserves further study. The following points may be the direction of efforts: (1) comprehensive analysis of the expression of different immune cells and immune factors in the same batch of samples. (2) The study should be conducted in different types of pituitary adenomas. (3) Further mechanism study based on the current apparent expression research. (4) Exploration of new immune cell types and immune factors. At the same time, the exploration of immunotherapy also provides a new choice for the treatment of PAs. It should also be kept in mind that the immune response is dynamic during tumor development and that there may be inter-patient heterogeneity. Individualized immunotherapy strategies for individual patients at different stages are very important. Therefore, it is necessary to determine the immune landscape and its mechanism changes in pituitary adenoma, which is undoubtedly a challenge for researchers.

Availability of data and materials

Not applicable.

Abbreviations

PAs:

Pituitary adenomas

TME:

Tumor microenvironment

NK:

Natural killer

ECM:

Extracellular matrix

TMICs:

Tumor microenvironmental immune cells

TLS:

Tertiary lymphoid structures

TAM:

Macrophages

DCs:

Dendritic cells

CTLS:

Cytotoxic T cells

FPAs:

Functional pituitary adenomas

NFPAs:

Non-functional pituitary adenomas

TIICs:

Tumor-infiltration immune cells

IFN:

Interferon

ILs:

Interleukin insurance-linked securities

TNF:

Tumor necrosis factor

MIF:

Macrophage migration inhibitory factor

CTLA-4:

Co-inhibitory cytotoxic T lymphocyte-associated protein 4

PD-1:

Programmed cell death 1

References

  1. Fabio G, et al. Pituitary adenoma and the chemokine network: a systemic view. Front Endocrinol. 2015;6:141.

    Google Scholar 

  2. Di Ieva A, et al. Aggressive pituitary adenomas—diagnosis and emerging treatments. Nat Rev Endocrinol. 2014;10(7):423–35. https://doi.org/10.1038/nrendo.2014.64.

    Article  CAS  PubMed  Google Scholar 

  3. Hansen TM, Batra S, Lim M, et al. Invasive adenoma and pituitary carcinoma: a SEER database analysis. Neurosurg Rev. 2014;37(2):279–86. https://doi.org/10.1007/s10143-014-0525-y.

  4. Heaney AP. Clinical review: Pituitary carcinoma: difficult diagnosis and treatment [published correction appears in J Clin Endocrinol Metab. 2012 Mar;97(3):1064]. J Clin Endocrinol Metab. 2011;96(12):3649–60. https://doi.org/10.1210/jc.2011-2031.

  5. Kemeny HR, Elsamadicy AA, Farber SH, Champion CD, Lorrey SJ, Chongsathidkiet P, et al. Targeting PD-L1 initiates effective antitumor immunity in a murine model of cushing disease. Clin Cancer Res. 2020;26(5):1141–51. https://doi.org/10.1158/1078-0432.CCR-18-3486.

    Article  CAS  PubMed  Google Scholar 

  6. Dai W, Zhuang Z, Ling H, Yang Y, Hang C. Systematic review and network meta-analysis assess the comparative efficacy and safety of transsphenoidal surgery for pituitary tumor. Neurosurg Rev. 2021;44(1):515–27. https://doi.org/10.1007/s10143-020-01240-3.

  7. Almutairi R, et al. Gross total resection of pituitary adenomas after endoscopic vs. microscopic transsphenoidal surgery: a meta-analysis. Acta Neurochir. 2018;160(5):1005–21. https://doi.org/10.1007/s00701-017-3438-z.

    Article  PubMed  Google Scholar 

  8. Lamb LS, Sim H-W, McCormack AI. Exploring the role of novel medical therapies for aggressive pituitary tumors: a review of the literature—“are we there yet?”. Cancers. 2020;12(2):308. https://doi.org/10.3390/cancers12020308.

    Article  PubMed Central  Google Scholar 

  9. Maghathe T, Miller WK, Mugge L, Mansour TR, Schroeder J. Immunotherapy and potential molecular targets for the treatment of pituitary adenomas resistant to standard therapy: a critical review of potential therapeutic targets and current developments. J Neurosurg Sci. 2020;64(1):71–83. https://doi.org/10.23736/S0390-5616.18.04419-3.

    Article  PubMed  Google Scholar 

  10. Suojun Z, Feng W, Dongsheng G, Ting L. Targeting Raf/MEK/ERK pathway in pituitary adenomas. Eur J Cancer. 2012;48(3):389–95. https://doi.org/10.1016/j.ejca.2011.11.002.

    Article  CAS  PubMed  Google Scholar 

  11. Strathmann FG, Borlee G, Born DE, Gonzalez-Cuyar LF, Huber BR, Baird GS. Multiplex immunoassays of peptide hormones extracted from formalin-fixed, paraffin-embedded tissue accurately subclassify pituitary adenomas. Clin Chem. 2012;58(2):366–74.

  12. Dragomir MP, Moisoiu V, Manaila R, et al. A holistic perspective: exosomes shuttle between nerves and immune cells in the tumor microenvironment. J Clin Med. 2020;9(11):3529. https://doi.org/10.3390/jcm9113529.

  13. Zhang J, Gu C, Song Q, Zhu M, Xu Y, Xiao M, et al. Identifying cancer-associated fibroblasts as emerging targets for hepatocellular carcinoma. Cell Biosci. 2020;10(1):127. https://doi.org/10.1186/s13578-020-00488-y.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  14. Marques P, Barry S, Carlsen E, Collier D, Ronaldson A, Awad S, et al. Chemokines modulate the tumour microenvironment in pituitary neuroendocrine tumours. Acta Neuropathol Commun. 2019;7(1):172. https://doi.org/10.1186/s40478-019-0830-3.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  15. Buchta Rosean CM. and M.R. Rutkowski, The influence of the commensal microbiota on distal tumor-promoting inflammation. Semin Immunol. 2017;32:62–73. https://doi.org/10.1016/j.smim.2017.06.002.

    Article  CAS  PubMed  Google Scholar 

  16. Hajjo H, Geva-Zatorsky N. Gut microbiota–host interactions now also brain-immune axis. Curr Opin Neurobiol. 2020;62:53–9. https://doi.org/10.1016/j.conb.2019.10.009.

    Article  CAS  PubMed  Google Scholar 

  17. Yong SB, Chung JY, Song Y, Kim J, Ra S, Kim YH. Non-viral nano-immunotherapeutics targeting tumor microenvironmental immune cells. Biomaterials. 2019;219:119401. https://doi.org/10.1016/j.biomaterials.2019.119401.

    Article  CAS  PubMed  Google Scholar 

  18. Belli C, Trapani D, Viale G, D'Amico P, Duso BA, Della Vigna P, et al. Targeting the microenvironment in solid tumors. Cancer Treat Rev. 2018;65:22–32. https://doi.org/10.1016/j.ctrv.2018.02.004.

    Article  CAS  PubMed  Google Scholar 

  19. Hui L, Chen Y. Tumor microenvironment: sanctuary of the devil. Cancer Lett. 2015;368(1):7–13. https://doi.org/10.1016/j.canlet.2015.07.039.

    Article  CAS  PubMed  Google Scholar 

  20. Ilie MD, Vasiljevic A, Raverot G, Bertolino P. The microenvironment of pituitary tumors—biological and therapeutic implications. Cancers. 2019;11(10):1605. https://doi.org/10.3390/cancers11101605.

    Article  CAS  PubMed Central  Google Scholar 

  21. Ramaswamy K. Human Immune System; 2003.

    Book  Google Scholar 

  22. Chiossone L, Dumas PY, Vienne M, Vivier E. Natural killer cells and other innate lymphoid cells in cancer. Nat Rev Immunol. 2018;18(11):671–88. https://doi.org/10.1038/s41577-018-0061-z.

    Article  CAS  Google Scholar 

  23. Malmberg KJ, Carlsten M, Björklund A, Sohlberg E, Bryceson YT, Ljunggren HG. Natural killer cell-mediated immunosurveillance of human cancer. Semin Immunol. 2017;31:20–9. https://doi.org/10.1016/j.smim.2017.08.002.

    Article  CAS  PubMed  Google Scholar 

  24. Ljunggren HG, Kärre K. In search of the ‘missing self’: MHC molecules and NK cell recognition. Immunol Today. 1990;11(7):237–44. https://doi.org/10.1016/0167-5699(90)90097-S.

    Article  CAS  PubMed  Google Scholar 

  25. Garrido F. MHC/HLA Class I Loss in Cancer Cells. Adv Exp Med Biol. 2019;1151:15–78. https://doi.org/10.1007/978-3-030-17864-2_2.

    Article  CAS  PubMed  Google Scholar 

  26. Dendrou CA, Petersen J, Rossjohn J, Fugger L. HLA variation and disease. Nat Rev Immunol. 2018;18(5):325–39. https://doi.org/10.1038/nri.2017.143.

    Article  CAS  PubMed  Google Scholar 

  27. Walzer T, Blery M, Chaix J, Fuseri N, Chasson L, Robbins SH, et al. Identification, activation, and selective in vivo ablation of mouse NK cells via NKp46. Proc Natl Acad Sci U S A. 2007;104(9):3384–9. https://doi.org/10.1073/pnas.0609692104.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  28. Weil S, et al. Natural killer group 2D ligand depletion reconstitutes natural killer cell immunosurveillance of head and neck squamous cell carcinoma. Front Immunol. 2017;8:387.

    Article  Google Scholar 

  29. Yang L, Zhang Y. Tumor-associated macrophages: from basic research to clinical application. J Hematol Oncol. 2017;10(1):58. https://doi.org/10.1186/s13045-017-0430-2.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  30. Grivennikov SI, Greten FR, Karin M. Immunity, inflammation, and cancer. Cell. 2010;140(6):883–99. https://doi.org/10.1016/j.cell.2010.01.025.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  31. Zhang A, Xu Y, Xu H, Ren J, Meng T, Ni Y, et al. Lactate-induced M2 polarization of tumor-associated macrophages promotes the invasion of pituitary adenoma by secreting CCL17. Theranostics. 2021;11(8):3839–52. https://doi.org/10.7150/thno.53749.

    Article  PubMed  PubMed Central  Google Scholar 

  32. Marques P, Barry S, Carlsen E, Collier D, Ronaldson A, Dorward N, et al. The role of the tumour microenvironment in the angiogenesis of pituitary tumours. Endocrine. 2020;70(3):593–606. https://doi.org/10.1007/s12020-020-02478-z.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  33. Hinshaw DC, Shevde LA. The tumor microenvironment innately modulates cancer progression. Cancer Res. 2019;79(18):4557–66. https://doi.org/10.1158/0008-5472.CAN-18-3962.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  34. Hashimoto M, Kamphorst AO, Im SJ, Kissick HT, Pillai RN, Ramalingam SS, et al. CD8 T cell exhaustion in chronic infection and cancer: opportunities for interventions. Annu Rev Med. 2018;69(1):301–18. https://doi.org/10.1146/annurev-med-012017-043208.

    Article  CAS  PubMed  Google Scholar 

  35. Tay RE, Richardson EK, Toh HC. Revisiting the role of CD4(+) T cells in cancer immunotherapy-new insights into old paradigms. Cancer Gene Ther. 2021;28(1-2):5–17. https://doi.org/10.1038/s41417-020-0183-x.

  36. Sadeghi Rad H, Monkman J, Warkiani ME, et al. Understanding the tumor microenvironment for effective immunotherapy. Med Res Rev. 2021;41(3):1474–98. https://doi.org/10.1002/med.2176.

  37. Zhang Y, Gallastegui N, Rosenblatt JD. Regulatory B cells in anti-tumor immunity. Int Immunol. 2015;27(10):521–30. https://doi.org/10.1093/intimm/dxv034.

    Article  CAS  PubMed  Google Scholar 

  38. Wang Z, Guo X, Gao L, Deng K, Lian W, Bao X, et al. The immune profile of pituitary adenomas and a novel immune classification for predicting immunotherapy responsiveness. J Clin Endocrinol Metab. 2020;105(9):e3207–23. https://doi.org/10.1210/clinem/dgaa449.

    Article  PubMed Central  Google Scholar 

  39. Aflorei E, Korbonits M. Epidemiology and etiopathogenesis of pituitary adenomas. J Neuro-Oncol. 2014;117(3):379–94. https://doi.org/10.1007/s11060-013-1354-5.

    Article  Google Scholar 

  40. Varlamov E, McCartney S, Fleseriu M. Functioning pituitary adenomas - current treatment options and emerging medical therapies. Eur Endocrinol. 2019;15(1):30–40. https://doi.org/10.17925/EE.2019.15.1.30.

    Article  PubMed  PubMed Central  Google Scholar 

  41. Yeung JT, Vesely MD, Miyagishima DF. In silico analysis of the immunological landscape of pituitary adenomas. J Neurooncol. 2020;147(3):595–8. https://doi.org/10.1007/s11060-020-03476-x.

    Article  PubMed  PubMed Central  Google Scholar 

  42. Wang PF, Wang TJ, Yang YK, Yao K, Li Z, Li YM, et al. The expression profile of PD-L1 and CD8(+) lymphocyte in pituitary adenomas indicating for immunotherapy. J Neurooncol. 2018;139(1):89–95. https://doi.org/10.1007/s11060-018-2844-2.

    Article  CAS  PubMed  Google Scholar 

  43. Salomon MP, Wang X, Marzese DM, Hsu SC, Nelson N, Zhang X, et al. The epigenomic landscape of pituitary adenomas reveals specific alterations and differentiates among acromegaly, Cushing's disease and endocrine-inactive subtypes. Clin Cancer Res. 2018;24(17):4126–36. https://doi.org/10.1158/1078-0432.CCR-17-2206.

    Article  CAS  PubMed  Google Scholar 

  44. Trouillas J, et al. A new prognostic clinicopathological classification of pituitary adenomas: a multicentric case–control study of 410 patients with 8 years post-operative follow-up. Acta Neuropathol. 2013;126(1):123–35. https://doi.org/10.1007/s00401-013-1084-y.

    Article  PubMed  Google Scholar 

  45. Fleseriu M, Popovic V. The journey in diagnosis and treatment, from pituitary adenoma to aggressive pituitary tumors. Rev Endocr Metab Disord. 2020;21(2):201–2. https://doi.org/10.1007/s11154-020-09561-w.

    Article  PubMed  Google Scholar 

  46. Principe, M., et al., Immune landscape of pituitary neuroendocrine tumours reveals association between macrophages and gonadotroph-tumour invasion. J Clin Endocrinol Metab, 2020.

  47. Principe M, Chanal M, Ilie MD, et al. Immune landscape of pituitary tumors reveals association between macrophages and gonadotroph tumor invasion. J Clin Endocrinol Metab. 2020;105(11):dgaa520. https://doi.org/10.1210/clinem/dgaa520.

  48. Lu JQ, Adam B, Jack AS, Lam A, Broad RW, Chik CL. Immune cell infiltrates in pituitary adenomas: more macrophages in larger adenomas and more T cells in growth hormone adenomas. Endocr Pathol. 2015;26(3):263–72. https://doi.org/10.1007/s12022-015-9383-6.

    Article  CAS  PubMed  Google Scholar 

  49. Mei Y, Bi WL, Greenwald NF, du Z, Agar NYR, Kaiser UB, et al. Increased expression of programmed death ligand 1 (PD-L1) in human pituitary tumors. Oncotarget. 2016;7(47):76565–76. https://doi.org/10.18632/oncotarget.12088.

    Article  PubMed  PubMed Central  Google Scholar 

  50. Sato M, Tamura R, Tamura H, et al. Analysis of tumor angiogenesis and immune microenvironment in non-functional pituitary endocrine tumors. J Clin Med. 2019;8(5):695. https://doi.org/10.3390/jcm8050695.

  51. Iacovazzo D, Chiloiro S, Carlsen E, Bianchi A, Giampietro A, Tartaglione T, et al. Tumour-infiltrating cytotoxic T lymphocytes in somatotroph pituitary neuroendocrine tumours. Endocrine. 2020;67(3):651–8. https://doi.org/10.1007/s12020-019-02145-y.

    Article  CAS  PubMed  Google Scholar 

  52. Zhou W, Zhang C, Zhang D, Peng J, Ma S, Wang X, et al. Comprehensive analysis of the immunological landscape of pituitary adenomas: implications of immunotherapy for pituitary adenomas. J Neurooncol. 2020;149(3):473–87. https://doi.org/10.1007/s11060-020-03636-z.

    Article  CAS  PubMed  Google Scholar 

  53. Barry S, Carlsen E, Marques P, Stiles CE, Gadaleta E, Berney DM, et al. Tumor microenvironment defines the invasive phenotype of AIP-mutation-positive pituitary tumors. Oncogene. 2019;38(27):5381–95. https://doi.org/10.1038/s41388-019-0779-5.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  54. Lupi I, Manetti L, Caturegli P, Menicagli M, Cosottini M, Iannelli A, et al. Tumor infiltrating lymphocytes but not serum pituitary antibodies are associated with poor clinical outcome after surgery in patients with pituitary adenoma. J Clin Endocrinol Metab. 2010;95(1):289–96. https://doi.org/10.1210/jc.2009-1583.

    Article  CAS  PubMed  Google Scholar 

  55. Gang, H., et al., The study on expression and relationship between matrix metalloproteinase-2 and CD147 in invasive pituitary adenomas. Chinese J Nervous Ment Dis. 2005.

  56. Zhao G, Chen W, He J, Cui C, Zhao L, Zhao Y, et al. Analysis of cyclooxygenase 2, programmed cell death ligand 1, and arginase 1 expression in human pituitary adenoma. World Neurosur. 2020;144:e660–73. https://doi.org/10.1016/j.wneu.2020.09.031.

    Article  Google Scholar 

  57. Rossi ML, Jones NR, Esiri MM, Havas L, Izzi MA, Coakham HB. Mononuclear cell infiltrate and HLA-Dr expression in 28 pituitary adenomas. Tumori. 1990;76(6):543–7. https://doi.org/10.1177/030089169007600605.

    Article  CAS  PubMed  Google Scholar 

  58. Briceno V, Zaidi HA, Doucette JA, Onomichi KB, Alreshidi A, Mekary RA, et al. Efficacy of transsphenoidal surgery in achieving biochemical cure of growth hormone-secreting pituitary adenomas among patients with cavernous sinus invasion: a systematic review and meta-analysis. Neurol Res. 2017;39(5):387–98. https://doi.org/10.1080/01616412.2017.1296653.

    Article  PubMed  Google Scholar 

  59. Ilie MD, Vasiljevic A, Raverot G, Bertolino P. The microenvironment of pituitary tumors-biological and therapeutic implications. Cancers (Basel). 2019;11(10):1605. https://doi.org/10.3390/cancers11101605.

  60. Qiu L, Yang J, Wang H, Zhu Y, Wang Y, Wu Q. Expression of T-helper-associated cytokines in the serum of pituitary adenoma patients preoperatively and postperatively. Med Hypotheses. 2013;80(6):781–6. https://doi.org/10.1016/j.mehy.2013.03.011.

    Article  CAS  PubMed  Google Scholar 

  61. Green VL, Atkin SL, Speirs V, Jeffreys RV, Landolt AM, Mathew B, et al. Cytokine expression in human anterior pituitary adenomas. Clin Endocrinol (Oxf). 1996;45(2):179–85. https://doi.org/10.1046/j.1365-2265.1996.d01-1554.x.

    Article  CAS  Google Scholar 

  62. Pestka S, Krause CD, Walter MR. Interferons, interferon-like cytokines, and their receptors. Immunol Rev. 2004;202(1):8–32. https://doi.org/10.1111/j.0105-2896.2004.00204.x.

    Article  CAS  PubMed  Google Scholar 

  63. Hofland LJ, de Herder WW, Waaijers M, Zuijderwijk J, Uitterlinden P, van Koetsveld P, et al. Interferon-alpha-2a is a potent inhibitor of hormone secretion by cultured human pituitary adenomas. J Clin Endocrinol Metab. 1999;84(9):3336–43. https://doi.org/10.1210/jcem.84.9.6005.

    Article  CAS  PubMed  Google Scholar 

  64. Gough P, Myles IA. Tumor necrosis factor receptors: pleiotropic signaling complexes and their differential effects. Front Immunol. 2020;11:585880. https://doi.org/10.3389/fimmu.2020.585880.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  65. Nanes MS. Tumor necrosis factor-alpha: molecular and cellular mechanisms in skeletal pathology. Gene. 2003;321:1–15. https://doi.org/10.1016/S0378-1119(03)00841-2.

    Article  CAS  PubMed  Google Scholar 

  66. Zhu H, Guo J, Shen Y, Dong W, Gao H, Miao Y, et al. Functions and mechanisms of tumor necrosis factor-α and noncoding RNAs in bone-invasive pituitary adenomas. Clin Cancer Res. 2018;24(22):5757–66. https://doi.org/10.1158/1078-0432.CCR-18-0472.

    Article  CAS  PubMed  Google Scholar 

  67. Xiao Z, Liu Q, Mao F, Wu J, Lei T. TNF-α-induced VEGF and MMP-9 expression promotes hemorrhagic transformation in pituitary adenomas. Int J Mol Sci. 2011;12(6):4165–79. https://doi.org/10.3390/ijms12064165.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  68. Glebauskiene B, et al. Association of Ki-67 Labelling Index and IL-17A with Pituitary Adenoma. Biomed Res Int. 2018;2018:7490585.

    Article  Google Scholar 

  69. Kalsi AK, Halder A, Jain M, Chaturvedi PK, Mathew M, Sharma JB. Association of raised levels of IL-4 and anti-TPO with hyperprolactinemia. Am J Reprod Immunol. 2019;81(3):e13085. https://doi.org/10.1111/aji.13085.

    Article  CAS  PubMed  Google Scholar 

  70. Sapochnik M, Fuertes M, Arzt E. Programmed cell senescence: role of IL-6 in the pituitary. J Mol Endocrinol. 2017;58(4):R241–r253. https://doi.org/10.1530/JME-17-0026.

    Article  CAS  PubMed  Google Scholar 

  71. Arzt E, Buric R, Stelzer G, Stalla J, Sauer J, Renner U, et al. Interleukin involvement in anterior pituitary cell growth regulation: effects of IL-2 and IL-6. Endocrinology. 1993;132(1):459–67. https://doi.org/10.1210/endo.132.1.8419142.

    Article  CAS  PubMed  Google Scholar 

  72. Takahashi N, Nishihira J, Sato Y, Kondo M, Ogawa H, Ohshima T, et al. Involvement of macrophage migration inhibitory factor (MIF) in the mechanism of tumor cell growth. Mol Med. 1998;4(11):707–14. https://doi.org/10.1007/BF03401765.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  73. Kishore C, Bhadra P. Bhadra, Current advancements and future perspectives of immunotherapy in colorectal cancer research. Eur J Pharmacol. 2021;893:173819. https://doi.org/10.1016/j.ejphar.2020.173819.

  74. Filin IY, Solovyeva VV, Kitaeva KV, Rutland CS, Rizvanov AA. Current trends in cancer immunotherapy. Biomedicines. 2020;8(12):621. https://doi.org/10.3390/biomedicines8120621.

  75. Dai C, Liang S, Sun B, Kang J. The Progress of Immunotherapy in Refractory Pituitary Adenomas and Pituitary Carcinomas. Front Endocrinol (Lausanne). 2020;11:608422. https://doi.org/10.3389/fendo.2020.608422.

    Article  Google Scholar 

  76. Sun Z, Fourcade J, Pagliano O, Chauvin JM, Sander C, Kirkwood JM, et al. IL10 and PD-1 Cooperate to Limit the Activity of Tumor-Specific CD8+ T Cells. Cancer Res. 2015;75(8):1635–44. https://doi.org/10.1158/0008-5472.CAN-14-3016.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  77. Wei SC, et al. Negative co-stimulation constrains T cell differentiation by imposing boundaries on possible cell states. Immunity. 2019;50(4):1084–1098.e10.

    Article  CAS  Google Scholar 

  78. Caturegli P, di Dalmazi G, Lombardi M, Grosso F, Larman HB, Larman T, et al. Hypophysitis secondary to cytotoxic T-lymphocyte-associated protein 4 blockade: insights into pathogenesis from an autopsy series. Am J Pathol. 2016;186(12):3225–35. https://doi.org/10.1016/j.ajpath.2016.08.020.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  79. Weiner DM, Durgin JS, Wysocka M, Rook AH. The immunopathogenesis and immunotherapy of cutaneous T cell lymphoma: part II, current and future approaches. J Am Acad Dermatol. 2021;84(3):597–604. https://doi.org/10.1016/j.jaad.2020.12.026.

  80. Tsushima F, Yao S, Shin T, Flies A, Flies S, Xu H, et al. Interaction between B7-H1 and PD-1 determines initiation and reversal of T-cell anergy. Blood. 2007;110(1):180–5. https://doi.org/10.1182/blood-2006-11-060087.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  81. Taube J, et al. Colocalization of inflammatory response with B7-h1 expression in human melanocytic lesions supports an adaptive resistance mechanism of immune escape. Sci Transl Med. 2012;4(127):127ra37.

    Article  Google Scholar 

  82. Pardoll DM. The blockade of immune checkpoints in cancer immunotherapy. Nat Rev Cancer. 2012;12(4):252–64. https://doi.org/10.1038/nrc3239.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  83. Fischli S, Allelein S, Zander T, Henzen C. Endocrinologic side effects of oncologic treatment with anti-CTLA-4-antibodies. Dtsch Med Wochenschr. 2014;139(19):996–1000. https://doi.org/10.1055/s-0034-1369961.

    Article  CAS  PubMed  Google Scholar 

  84. Sharma P, Allison JP. Dissecting the mechanisms of immune checkpoint therapy. Nat Rev Immunol. 2020;20(2):75–6. https://doi.org/10.1038/s41577-020-0275-8.

    Article  CAS  PubMed  Google Scholar 

  85. Kim DY, Kim PH, Suh CH, Kim KW, Kim HS. Immune checkpoint inhibitors with or without radiotherapy in non-small cell lung cancer patients with brain metastases: a systematic review and meta-analysis. Diagnostics (Basel). 2020;10(12):1098.

  86. Sato M, Tamura R, Tamura H, et al. Analysis of tumor angiogenesis and immune microenvironment in non-functional pituitary endocrine tumors. J Clin Med. 2019;8(5):695. https://doi.org/10.3390/jcm8050695.

  87. Sol B, de Filette JMK, Awada G, Raeymaeckers S, Aspeslagh S, Andreescu CE, et al. Immune checkpoint inhibitor therapy for ACTH-secreting pituitary carcinoma: a new emerging treatment? Eur J Endocrinol. 2021;184(1):K1–k5. https://doi.org/10.1530/EJE-20-0151.

    Article  CAS  PubMed  Google Scholar 

  88. Xu X, Wang D, Wu W, Lu H. Molecular characteristics of genes and the immune microenvironment of a rare chest malignant tumor (pulmonary clear cell sarcoma): a case report. Front Oncol. 2021;11:664883. https://doi.org/10.3389/fonc.2021.664883.

    Article  PubMed  PubMed Central  Google Scholar 

  89. Hazrati SM, Aghazadeh J, Mohtarami F, Abouzari M, Rashidi A. Immunotherapy of prolactinoma with a T helper 1 activator adjuvant and autoantigens: a case report. Neuroimmunomodulation. 2006;13(4):205–8. https://doi.org/10.1159/000100405.

    Article  CAS  PubMed  Google Scholar 

  90. Lakomý R, Poprach A, Kazda T. Management of immune-related endocrinopathies in anticancer treatment with checkpoint inhibitors. Klin Onkol. 2020;33(1):15–9. https://doi.org/10.14735/amko202015.

    Article  PubMed  Google Scholar 

  91. Castinetti F, Albarel F, Archambeaud F, Bertherat J, Bouillet B, Buffier P, et al. French Endocrine Society Guidance on endocrine side effects of immunotherapy. Endocrine-related Cancer. 2019;26(2):G1–G18. https://doi.org/10.1530/ERC-18-0320.

    Article  CAS  PubMed  Google Scholar 

  92. Deng L, Liang H, Burnette B, Beckett M, Darga T, Weichselbaum RR, et al. Irradiation and anti–PD-L1 treatment synergistically promote antitumor immunity in mice. J Clin Invest. 2014;124(2):687–95. https://doi.org/10.1172/JCI67313.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

Download references

Acknowledgements

None.

Funding

No funding.

Author information

Authors and Affiliations

Authors

Contributions

Ding Nie and Qiuyue Fang carried out the studies. Ding Nie participated in collecting the data and drafted the manuscript. Jianhua Cheng and Bin Li analyzed the data. Chuzhong Li and Songbai Gui directed the writing. Yazhuo Zhang and Peng Zhao helped to revise the manuscript. The authors read and approved the final manuscript.

Corresponding author

Correspondence to Peng Zhao.

Ethics declarations

Ethics approval and consent to participate

Not applicable.

Consent for publication

Not applicable.

Competing interests

The authors declare that they have no competing interests.

Additional information

Publisher’s Note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Rights and permissions

Open Access This article is licensed under a Creative Commons Attribution 4.0 International License, which permits use, sharing, adaptation, distribution and reproduction in any medium or format, as long as you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons licence, and indicate if changes were made. The images or other third party material in this article are included in the article's Creative Commons licence, unless indicated otherwise in a credit line to the material. If material is not included in the article's Creative Commons licence and your intended use is not permitted by statutory regulation or exceeds the permitted use, you will need to obtain permission directly from the copyright holder. To view a copy of this licence, visit http://creativecommons.org/licenses/by/4.0/. The Creative Commons Public Domain Dedication waiver (http://creativecommons.org/publicdomain/zero/1.0/) applies to the data made available in this article, unless otherwise stated in a credit line to the data.

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Nie, D., Fang, Q., Li, B. et al. Research advances on the immune research and prospect of immunotherapy in pituitary adenomas. World J Surg Onc 19, 162 (2021). https://doi.org/10.1186/s12957-021-02272-9

Download citation

  • Received:

  • Accepted:

  • Published:

  • DOI: https://doi.org/10.1186/s12957-021-02272-9