Next Article in Journal
The Interplay of Perceived Risks and Benefits in Deciding to Become Vaccinated against COVID-19 While Pregnant or Breastfeeding: A Cross-Sectional Study in Italy
Previous Article in Journal
Transfusion Burden in Allogeneic Hematopoietic Stem Cell Transplantation over Time: Experience from a Single Institution
Previous Article in Special Issue
Cognitive Dysfunction, an Increasingly Valued Long-Term Impairment in Acromegaly
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Progress and Challenges of Immune Checkpoint Inhibitor-Induced Hypophysitis

1
Department of Endocrinology and Metabolism, West China Hospital, Sichuan University, Chengdu 610041, China
2
Institute of Pituitary Adenomas and Related Diseases, West China Hospital, Sichuan University, Chengdu 610041, China
*
Authors to whom correspondence should be addressed.
J. Clin. Med. 2023, 12(10), 3468; https://doi.org/10.3390/jcm12103468
Submission received: 31 December 2022 / Revised: 13 April 2023 / Accepted: 29 April 2023 / Published: 15 May 2023
(This article belongs to the Special Issue Pituitary Tumors: Diagnosis and Treatment)

Abstract

:
Immune checkpoint inhibitors (ICIs) are a new type of antitumor drug which can achieve antitumor goals by blocking the binding of immune checkpoints to their ligands, thereby enhancing the activity of T cells. Meanwhile, ICIs block the binding of immune checkpoints to their ligands, disrupting the immune tolerance of T cells to self-antigens, which may lead to a series of immune-related adverse events (irAEs). Immune checkpoint inhibitor-induced hypophysitis (IH) is a relatively rare irAE. Due to the lack of specificity in clinical manifestations, it is difficult to accurately diagnose IH in a timely manner in clinical practice. However, the risk of adverse events, especially IH, for patients receiving ICIs has not been adequately investigated. Missed or delayed diagnosis may lead to poor prognosis or even adverse clinical outcomes. In this article, we summarize the epidemiology, pathogenesis, clinical manifestations, diagnosis and treatment of IH.

1. Definition and Classification of ICIs

Malignant tumors are a major disease that seriously threatens human health. Surgery, radiotherapy and chemotherapy are the main means of tumor treatment. Targeted therapy and immunotherapy are new methods of tumor treatment that have been developed in recent years and are revolutionary breakthroughs in tumor treatment. Among them, immune checkpoint inhibitors (ICIs) are the main methods of current immunotherapy. Immune checkpoints are one normal part of the immune system. ICIs work by blocking checkpoint proteins from binding with their partner proteins. However, ICIs lack tumor tissue specificity and destroy the immune tolerance of T cells to self-antigens, which can lead to a series of immune-related adverse events (irAEs) [1]. As a common endocrine irAE, immune checkpoint inhibitor-induced hypophysitis (IH) cannot be diagnosed in a timely and accurate manner because of its lack of specificity in clinical manifestations, such as anterior pituitary insufficiency. Therefore, clinicians need to be vigilant enough in the process of tumor immunotherapy. On the one hand, the diagnosis and treatment of IH in a timely manner can avoid pituitary crises or even life-threatening situations. On the other hand, misdiagnosis of IH may lead to clinical decisions to discontinue immunotherapy, which may lead to tumor progression [2]. IH has brought new challenges to tumor treatment. Therefore, in this review, we summarize the epidemiology, pathogenesis, clinical manifestations, diagnosis and treatment of IH.
Immune checkpoints are small molecules expressed on the surface of T lymphocytes and play an important role in maintaining immune homeostasis and autoimmune tolerance.
There are two broad categories of immune checkpoints. One type of immune checkpoint, such as CD28 and CD27, enhances T-cell activity by mediating stimulatory signals; the other type of immune checkpoint mainly includes cytotoxic T-lymphocyte antigen 4 (CTLA-4) and programmed death protein 1 (PD-1), which blunts T cells by transmitting inhibitory signals and plays an important role in immune tolerance.
CTLA-4 acts at the initial stage of the immune response and can transmit T-cell inhibitory signals after binding to its ligands CD80 and CD86 [3]. In contrast, PD-1 generally acts in the later stage of the immune response and inhibits the activity and immune function of T cells after binding to its ligands’ programmed death receptor ligand 1/2 (PD-L1/2) [4,5]. Tumor cells evade the body’s immune attack by enhancing the inhibitory signals of CTLA-4 and PD-1 [3]. ICIs are a class of specific IgG antibodies that recognize the inhibitory immune checkpoints CTLA-4 or PD-1 [6] and immune checkpoint ligand PD-L1/2, which restore or enhance the killing power of T cells against tumor cells by blocking the interaction with their ligands to achieve antitumor effects [7]. Currently, ICIs mainly include CTLA-4 inhibitors and PD-1/PD-L1 inhibitors. To date, two CTLA-4 inhibitors, ipilimumab and tremelimumab, have been approved for clinical use in the European Union and the Food and Drug Administration (FDA) has approved them in the U.S., mainly for the treatment of melanoma. The PD-1 inhibitors that have been approved mainly include pembrolizumab, nivolumab and cemiplimab, which are mainly used for the treatment of malignant tumors such as melanoma, non-small-cell lung cancer (NSCLC), renal cancer, Hodgkin lymphoma, head and neck squamous cell carcinoma and urothelial carcinoma. There are also four types of PD-1 inhibitors developed independently, namely toripalimab, sintilimab, camrelizumab and tislelizumab, all of which have been approved by the State Food and Drug Administration (SFDA) in China and are mainly approved for the treatment of Hodgkin lymphoma. PD-L1 inhibitors mainly include atezolizumab, avelumab and durvalumab, which are mainly used for the treatment of urothelial cancer and NSCLC. To expand the clinical indications of the above ICIs for more tumors, many preclinical studies are underway.
Accessed on 30 June 2022, there are a total of 616 ongoing clinical studies of CTLA-4 inhibitors and a total of 4890 ongoing clinical studies of PD-1/PD-L1 inhibitors registered in the U.S. National Library of Medicine’s official website of clinical trials (https://clinicaltrials.gov/, accessed on 30 June 2022) and China’s drug clinical trial information and publicity platform (http://www.chinadrugtrials.org.cn, accessed on 30 June 2022). It is expected that the clinical indications of these two types of ICIs will further increase in the near future, and the clinical application will become increasingly extensive. The representative drugs of CTLA-4 monoclonal antibody and PD-1/PD-L1 monoclonal antibody that have been approved for marketing, their time of approval and their detailed clinical indications are shown in Table 1.

2. Overview of irAEs

ICIs disrupt the maintenance of the body’s autoimmune tolerance, leading to a series of irAEs through multiple pathways, including autoreactive T cells, autoantibodies and cytokines. IrAEs can involve multiple organ systems, most commonly in the skin and gastrointestinal tract, followed by the lungs, blood, urinary, nervous and musculoskeletal systems, as well as cardiovascular systems [8]. The endocrine system is one of the most easily involved systems by irAEs due to its rich blood supply [9].
Since the European Society for Medical Oncology (ESMO) [10] published the world’s first clinical practice guideline on the management of irAEs in 2017, the American Society for Immunotherapy [8], the American Society of Clinical Oncology (ASCO), the National Comprehensive Cancer Network (NCCN) [11] and the Chinese Society of Clinical Oncology (CSCO) have successively issued clinical guidelines for the management of irAEs.
Since 2018, the British Endocrine Society [12], the Japanese Endocrine Society [13] and the French Endocrine Society [14] have successively published guidelines for the management of adverse endocrine reactions related to ICIs therapy. In 2021, The Chinese Society of Endocrinology (CSE) and the Professional Committee of Oncology and Endocrinology of the Chinese Anti-Cancer Association will also successively publish two expert consensuses on irAEs of the endocrine system caused by ICIs [15,16]. Endocrine-system-related irAEs have been reported thus far, including immunotherapy-related diabetes, thyroid dysfunction, adrenal insufficiency and IH [17]. Although the overall incidence of IH is not high, it has gradually attracted the attention of clinicians and scholars all over the world [18,19,20]. Due to IH’s lack of specificity in clinical manifestations, misdiagnosis and delayed diagnosis lead to poor prognosis for patients.
The abovementioned expert consensus and clinical guidelines on irAEs of the endocrine system have explained the diagnosis and treatment of IH. Correct diagnosis and timely treatment of IH is very important for the prognosis of tumor patients.

3. Epidemiology of ICI-Induced Hypophysitis

The first case of IH was reported by Phan et al. in 2003 [21]. A 54-year-old man with melanoma developed personality changes and memory loss after four cycles of treatment with the CTLA-4 inhibitor ipilimumab. During the 5th cycle of treatment, laboratory tests showed that the patient’s anterior pituitary dysfunction, thyroid-stimulating hormone (TSH), free thyroxine (FT4), adrenocorticotrophic hormone (ACTH), cortisol, growth hormone (GH), prolactin (PRL) and testosterone (T) levels were all undetectable. His saddle MRI showed a plump pituitary gland. The patient was diagnosed with IH, and his clinical symptoms improved following hydrocortisone, thyroxine and testosterone replacement therapy.
In 2016, Ishikawa et al. [22] reported a case of PD-1 inhibitor-related IH in a 55-year-old male Japanese patient with melanoma and right adrenal metastasis. The patient developed nausea, anorexia and weight loss after four cycles of nivolumab treatment. Laboratory tests showed that ACTH and cortisol decreased significantly, and the symptoms gradually improved after hydrocortisone treatment. Therefore, the patient was diagnosed with nivolumab-related IH. Since then, IH has gradually been reported and has received more attention from clinicians [17,23,24].
IH is a common endocrine-system-related irAE. The incidence of IH is related to the type and treatment regimen of ICI [25]. To date, multiple systematic reviews have conducted statistical analysis on the incidence of IH [2,18,19,20,26,27,28,29,30,31,32,33], and the results show that the combination of CTLA-4 inhibitors and PD-1/PD-L1 inhibitors has the highest incidence of IH. It constitutes up to 10% of patients who received treatment [31]. The incidence of IH associated with CTLA-4 inhibitor and PD-1/PD-L1 inhibitor monotherapy is much lower than that of combination therapy, accounting for 5% and 1%, respectively [26,29]. A summary of IH reports in ICI-related adverse reactions is shown in Table 2.
With the development of ICI clinical trials and increasing clinical application, the incidence statistics of IH may change. The incidence of IH associated with the CTLA-4 inhibitor ipilimumab has been reported to increase with increasing drug dose [34], and the incidence of IH was approximately 1.8–3.3% in patients receiving low-dose ipilimumab (<3 mg/kg). When the ipilimumab dose was increased (>3 mg/kg), the incidence of IH reached 9–17% [9]. Currently, there are few reports on PD-1/PD-L1 inhibitor-related IH, so the relationship between the incidence of PD-1/PD-L1 inhibitor-related IH and dose is still inconclusive. Although studies have shown no gender difference in irAEs [35], CTLA-4 inhibitor-related IH mostly occurs in men around the age of 60 [4], with a male-to-female ratio of about 4:1 [36]. Male predominance may be because the CTLA-4 inhibitor ipilimumab is commonly used in the treatment of melanoma, and men have a higher incidence of melanoma than women [25]. The median age of onset of PD-1/PD-L1 inhibitor-related IH is between 60 and 65 years old, and the male-to-female ratio is about 1–3:1 [37,38]. The data were derived from two follow-up studies involving 17 and 22 patients with PD-1 inhibitor-induced IH. Due to the limited number of patients in the two studies, whether there is a sex difference in PD-1/PD-L1 inhibitor-related IH still needs to be further supported by clinical data. Recent studies have found that the occurrence time of IH is related to the type of ICI [37,38,39,40]. IH associated with combination therapy of CTLA-4 inhibitor and PD-1/PD-L1 inhibitor generally occurs approximately 4 weeks after treatment. CTLA-4 inhibitor monotherapy mostly occurs approximately 9.3 weeks after the initial treatment [38]. PD-1/PD-L1 inhibitor-related IH occurs relatively late, with a median onset time of approximately 28 weeks [37], and IH has even been reported to occur approximately 60 weeks after discontinuation of PD-1/PD-L1 inhibitors [41].
In conclusion, male patients currently receiving CTLA-4 inhibitor and PD-1/PD-L1 inhibitor combination therapy have the highest incidence of IH, followed by CTLA-4 inhibitor monotherapy, and there may be a dose-dependent relationship. The lowest incidence was in patients receiving PD-1/PD-L1 inhibitor monotherapy. The onset time ranged from 4 weeks to 28 weeks after receiving ICI treatment, and some even set on after the drug was discontinued. The relevant epidemiological data of IH still need to be supplemented and improved by more clinical studies in the future.

4. Pathogenesis of ICI-Induced Hypophysitis

The exact pathogenesis that induces irAEs is still not clear. It was believed that its pathophysiological mechanism is related to the imbalance of immune checkpoints in maintaining immune system homeostasis. At present, some progress has been made in research on the mechanism of IH. Iwama et al. [42] confirmed the expression of CTLA-4 antigen in the pituitary tissue of a mouse hypophysitis model induced by a CTLA-4 inhibitor in animal experiments. An autopsy of patients who died after IH revealed that their pituitary tissue expressed CTLA-4, especially on pituitary TSH and PRL cells [42,43], suggesting that CTLA-4 inhibitors can directly bind to CTLA-4 on pituitary cells and degrade it as an antigen presented to CD8+ T cells, eventually inducing IH through the type IV hypersensitivity system [44]. This is consistent with the clinical manifestations that CTLA-4 inhibitor-related IH is prone to impaired thyroid axis function. In addition, among CTLA-4 inhibitors, the IgG subtype of ipilimumab is IgG1, while the IgG subtype of tremelimumab is IgG2 [42]. Both IgG subtypes can trigger antibody-dependent cell-mediated cytotoxicity (ADCC) and the classical complement activation pathway of type II hypersensitivity reactions [45]. Thus, CTLA-4 inhibitors can induce IH through type II hypersensitivity reactions.
Compared with CTLA-4 inhibitors, the mechanism of PD-1/PD-L1 inhibitor-induced IH is still unknown. Among them, PD-LI inhibitors are also of the IgG1 subtype, but their Fc fragments are modified, so they cannot induce IH through type II hypersensitivity reactions [46]. Since the main components of PD-1 inhibitors are IgG4-like IgG-4κ or IgG4 monoclonal antibodies, Chinese scholars speculate that PD-1 inhibitor-related IH may be similar to the pathogenesis of IgG4-related hypophysitis [47]. However, in fact, the specific pathogenesis of IgG4-related hypophysitis still has not been fully elucidated [48]. Antipituitary antibodies are present in the serum of patients with IgG4-related hypophysitis. The researchers treated the biopsied pituitary tissue with these antipituitary antibodies and found that growth hormone and opioid melanin may be the suspected self-antigens [49,50]. Keitaro Kanie et al. [51] used a similar method to process serum and tumor specimens from 20 patients with IH after treatment with PD-1/PD-L1 inhibitors. The results show the presence of antipituitary antibodies, anticorticotropin antibodies and anti-GH antibodies in the peripheral blood of these IH patients. There are antipituitary antibodies or antipituitary hormone autoantibodies both in the peripheral blood of patients with IgG4-related hypophysitis and PD-1/PD-L1 inhibitor-related IH, suggesting that PD-1/PD-L1 inhibitor-related IH and IgG4-related hypophysitis may have similar pathogenesis. Autoimmunity is involved, but the specific pathogenesis needs to be further explored. In addition, Tahir et al. [52] used the serological analysis of a recombinantly expressed cDNA clone (SEREX) to screen for autoantibodies in serum samples from patients with IH induced by the CTLA-4 inhibitor ipilimumab, the PD-1 inhibitor nivolumab, or a combination of two ICIs, which revealed that antiguanine nucleotide-binding protein G subunit alpha (GNAL) and anti-integral membrane protein 2B (ITM2B) autoantibodies were significantly increased. GNAL and ITM2B are proteins expressed in normal brain tissue [53,54]. Therefore, it is speculated that anti-GNAL and anti-ITM2B antibodies may be potential risk factors for the occurrence of IH [52].
In short, CTLA-4 inhibitors induce IH mainly through type II and type IV hypersensitivity reactions, and the pathogenesis of PD-1/PD-L1 inhibitor-related IH may be similar to IgG4-related hypophysitis. Anti-GNAL and anti-ITM2B antibodies may also potentially be involved in IH. However, the exact pathogenesis of IH remains to be further explored.

5. Clinical Features of ICI-Induced Hypophysitis

The clinical presentation of patients with IH often lacks specificity. In general, headache, fatigue, loss of appetite, and an enlarged pituitary gland are the main features, while central diabetes insipidus and visual disturbances are relatively rare. There are certain differences in specific performance due to different types of ICIs [44].
CTLA-4 inhibitor-related IH commonly manifests as headache and fatigue [38], and panpituitarism is often accompanied by pituitary enlargement [38]. IH associated with PD-1/PD-L1 inhibitors often presents with fatigue and poor appetite [37] and is often characterized by isolated secondary adrenal insufficiency [37,55]. According to the literature reports, the two types of ICI-related IH have different degrees of involvement in anterior pituitary function. The most common presentations of CTLA-4 inhibitor-related IH are FSH and/or LH deficiency (88%) and TSH deficiency (81%), followed by ACTH deficiency (55%) [38]. PD-1/PD-L1 inhibitor-related IH has ACTH deficiency in nearly 100% of cases [37], while TSH, GH and FSH/LH deficiency are less common, and its incidences were 11.8%, 13.3% and 18.8% of the treated population, respectively [37]. Most important of all is that the clinical manifestations of ACTH deficiency sometimes are the most severe [23,56,57]. Severe IH patients may present with high fever, low blood pressure, electrolyte disturbances (mainly hyponatremia) and even pituitary crises, such as disturbance of consciousness and coma. Since the symptoms of IH are not specific, they often need to be identified with sepsis with similar manifestations [15]. Central diabetes insipidus due to posterior pituitary involvement in IH is rare. To date, only a few reports of central diabetes insipidus have been reported. Among them, two cases were caused by the CTLA-4 inhibitor ipilimumab [58,59], two cases were caused by the PD-1 inhibitor nivolumab [60,61], and one case was caused by the PD-L1 inhibitor avelumab [62]. Notably, this type of IH-related hyponatremia needs to be differentiated from the syndrome of inappropriate antidiuretic hormone secretion (SIADH) caused by the tumor.
Pituitary or sellar imaging is helpful for differentiating IH from pituitary tumor metastases, pituitary adenomas and infectious pituitary diseases. Among them, pituitary-enhanced MRI is a more sensitive method. In total, 98% of patients with CTLA-4 inhibitor-related IH had pituitary enlargement [38], while only 28% of patients with PD-1/PD-L1 inhibitor-related IH developed pituitary enlargement [38]. A longitudinal retrospective study suggested that imaging changes in the pituitary can occur before or after the onset of clinical symptoms and changes in hormone levels [36]. Therefore, the absence of significant pituitary growth in early pituitary-enhanced MRI does not rule out IH [4,23]. The differences in clinical manifestations, laboratory tests and pituitary MRI between CTLA-4 inhibitor and PD-1/PD-L1 inhibitor-related IH are summarized in Table 3.
The symptoms of IH present with anterior pituitary hypofunction and imaging changes in the pituitary, which lack clinical specificity. Therefore, IH is easily missed or delayed in diagnosis. However, laboratory tests are sensitive and useful for diagnosing IH. Castinetti and Albarel et al. [63,64] suggested that pituitary-related hormones and pituitary MRI should be evaluated before using ICIs. Pituitary-related hormones include TSH, FT3 and FT4 in the hypothalamic–pituitary–thyroid axis (HPT axis), ACTH and cortisol in the hypothalamic–pituitary–adrenal axis (HPA axis), as well as ACTH stimulation test if necessary, and FSH, LH and T/E2 in the hypothalamic–pituitary–gonad axis (HPG axis). In addition, assessments of GH, IGF-1 and PRL are also useful. Meanwhile, indicators such as serum electrolytes, blood osmolality, urine osmolality and urine-specific gravity need to be measured to evaluate posterior pituitary function. According to the median onset time of IH, ClaireBriet et al. [39] suggested that during the first 6 months of ICI treatment, anterior pituitary function and blood electrolytes should be tested in each course of treatment for patients without symptoms of hypopituitarism. During the second 6 months of treatment, assessments are recommended every 2 courses. After 12 months of treatment with ICIs, pituitary hormone assessments are no longer routinely performed, because the usual time of onset of IH has passed (Figure 1). However, if the patient develops suspicious symptoms such as headache, fatigue, loss of appetite, etc., it is still necessary to evaluate anterior pituitary function and blood electrolytes. Pituitary imaging is not required in the assessment of anterior pituitary function described above; however, if anterior pituitary hypofunction is present, pituitary imaging is needed. Therefore, in the process of using ICIs, if the patient has clinical symptoms of suspected anterior pituitary hypofunction, anterior and posterior pituitary function should be evaluated immediately, and pituitary imaging examination should be completed at the same time [56].

6. Diagnosis of ICI-Induced Hypophysitis

At present, there is no consistent diagnostic standard for IH in the guidelines of various countries or regions around the world [14,39], but they all recommend that when patients using ICIs have clinical symptoms of suspected anterior pituitary dysfunction (such as fatigue, decreased appetite, etc.) and/or hyponatremia and/or examination suggests ≥1 anterior pituitary hormone deficiency (essential TSH or ACTH deficiency) and/or abnormal pituitary imaging findings, IH should be considered. In 2021, the expert consensus of the Chinese Society of Endocrinology and the Professional Committee of Oncology and Endocrinology of the Chinese Anti-Cancer Association recommended that the diagnostic criteria for IH be that patients develop more than one anterior pituitary hormone deficiency (must have TSH or ACTH deficiency) after using ICIs, accompanied by abnormal pituitary MRI performance or that patients develop more than two anterior pituitary hormone deficiencies (must have TSH or ACTH deficiency) and headache, as well as other symptoms of anterior pituitary hypofunction [15,16]. However, this criterion may fail to diagnose some patients with PD-1/PD-L1 inhibitor-related IH, whose main manifestation is isolated hypoadrenalism without pituitary MRI abnormalities [38]. Therefore, special vigilance should be paid to such patients without pituitary MRI abnormalities.
The severity of IH is different, and the follow-up diagnosis and treatment are also different. The “CSCO management guidelines on toxicities from immune checkpoint inhibitors” issued by the Chinese Society of Clinical Oncology (CSCO), as well as the “Chinese expert consensus on the immune checkpoint inhibitors-induced endocrine immune-related adverse events (2020)” issued by the Immunological Endocrinology Group, Endocrinology Society and Chinese Medical Association [15], all recommended the common terminology criteria for adverse events (CTCAE) published by the National Cancer Institute (NCI) to describe the severity of IH.
The severity of IH is graded from 1 to 5. Grade 1 means mild (no symptoms or mild symptoms), and Grade 2 means moderate (mild-to-moderate and mild local symptoms; noninvasive treatment needed; mild impairment of self-care; and no limitation of daily activities). Grade 3 means severe (severe or clinically important, but not temporarily life-threatening; requiring or prolonged hospitalization; severely impaired self-care; and affecting daily activities). Grade 4 means life-threatening (life-threatening, requiring urgent intervention; unable to perform daily activities). Grade 5 means death (Table 4). At present, IHs of CTCAE grades 1 to 5 have been reported, but most of them are CTCAE grades 1 to 2. Severe IHs (CTCAE grades 3 to 5) are relatively rare, accounting for approximately 10% to 40% of all cases of IH [18,19,20].

7. Treatment and Prognosis of ICI-Induced Hypophysitis

For the treatment and prognosis of IH, the British Endocrine Society [12], the Chinese Endocrinology Society [15] and the Chinese Anti-Cancer Association Tumor Endocrinology Professional Committee [16] all recommended that the severity of IH should be graded by CTCAE, according to the clinical manifestations of patients. Meanwhile, laboratory tests, such as anterior pituitary hormones, blood electrolytes, blood osmolality, urine osmolality and urine specific gravity, are necessary.
For IH patients assessed as CTCAE Grade 1 (mild fatigue, anorexia or mood changes without headache or asymptomatic), the ESMO and Chinese expert consensus suggests that ICI treatment can be continued. At the same time, according to the results of laboratory evaluation, the corresponding hormone replacement therapy should be performed for the damaged pituitary-target gland axis [16,65]. For CTCAE Grade 2 IH patients (headache without visual disturbance), ESMO recommends that ICI therapy should be suspended, and prednisone at 0.5–1 mg/kg/d should be administered orally [65]. If the patient’s symptoms do not relieve within 48 h, intravenous methylprednisolone at 0.5–1 mg/kg/d should be given, and the dose of prednisone should be reduced to 5 mg/d oral maintenance therapy within 2–4 weeks [65]. The damaged pituitary-target gland axis should be treated with corresponding hormone replacement therapy. Since discontinuation of ICIs does not affect the natural history of hypophysitis [66], and the survival benefit of continuation of ICI treatment of tumors is far greater than the risk of anterior pituitary dysfunction, ESMO recommends that patients with IH continue to use ICIs after the acute phase [65]. The expert consensus of the Chinese Cancer Society [15,16] proposed that ICI treatment should not be interrupted for CTCAE Grade 2 IH. For patients with IH in CTCAE Grades 3–4 (severe mass effect or severe headache; visual disturbance; or severe adrenal insufficiency or hypotension; or severe electrolyte disturbance), all guidelines recommend suspending ICI therapy [67] and immediately starting intravenous methylprednisolone at 1 mg/kg/d, as well as reducing glucocorticoids to prednisone at 5 mg/d oral maintenance therapy within 2–4 weeks. At the same time, according to the test results, other damaged pituitary–target gland axes were given corresponding hormone replacement therapy. Until the acute phase of IH is relieved, ICI treatment is continued [65]. For patients with ACTH and TSH deficiency, the Japan Endocrine Society [13] recommends that low-dose levothyroxine (12.5–25 µg/d) be administered after hydrocortisone (10–20 mg/d) for 5–7 d, adjusting the levothyroxine dose according to the serum level of FT4 to avoid inducing iatrogenic adrenal crisis. Studies have found that receiving high-dose (average daily prednisone dose greater than 7.5 mg) glucocorticoid therapy will affect the clinical antitumor efficacy of ICIs, reduce the overall survival rate of patients [68], increase the risk of infection, hyperglycemia, etc. [66], and cannot significantly improve the prognosis of anterior pituitary dysfunction [11]. In summary, high-dose glucocorticoid therapy is not recommended [13,15]. However, for patients with intractable headache and/or visual impairment, the Japanese Endocrine Society recommends the use of prednisone at 0.5–1.0 mg/kg/d [13]. Once symptoms such as intractable headaches and/or visual disturbances resolve, glucocorticoids should be rapidly reduced to physiologic replacement doses within 2–4 weeks [65]. Since HPG axis damage is easier to recover and poses a lower threat to the survival of patients, the French Endocrine Association [14] suggested that patients with HPG axis damage should be followed up for three months. If the HPG axis has not recovered after three months, the corresponding sex hormone replacement therapy can be given according to the patient’s age and other circumstances. For patients with an impaired GH/IGF-1 axis, the French Society of Endocrinology does not recommend GH replacement therapy due to the background of the patient’s primary malignancy [14]. If the posterior pituitary is damaged and diabetes insipidus occurs, the Chinese Society of Endocrinology recommends the use of synthetic antidiuretic hormone (ADH) for treatment [15]. Based on the recommendations of many guidelines, this review article briefly summarizes the diagnosis and treatment process of IH (Figure 1).
Compared with other irAEs, the prognosis of IH is relatively good. Generally, after the acute phase of the disease, the treatment of ICIs can be resumed. Permanent HPA axis damage occurs in 86–100% of IH patients, requiring long-term glucocorticoid replacement therapy [69]. Permanent HPA axis damage in PD-1/PD-L1 inhibitor-related IH is more common [37]. Today, with the increasing use of PD-1/PD-L1, it is especially necessary to attract the attention of clinicians. In addition, studies have shown that the HPT axis often recovers at approximately 10.5 weeks, and the HPG axis can recover at approximately 25 weeks [66]. However, 13–36% of patients may have permanent damage to the HPG axis [66,69]. Since the degree of damage and the recovery time to the abovementioned pituitary-target gland axis is not synchronized [15], the French Endocrine Society [14] recommends that from the first 6 months of the diagnosis of IH, the pituitary-target gland axis hormones need to be evaluated before each course of treatment to facilitate the timely detection of new pituitary-target gland axis damage. After 6 months, the pituitary-target gland axis hormones were evaluated every 3 months. After 12 months, the pituitary-target gland axis hormones were evaluated twice per year. Meanwhile, it is recommended to review the pituitary MRI every 3 months after the diagnosis of IH, which can evaluate the progress of IH and help rule out pituitary metastases from the primary tumor (Figure 1).

8. Summary

In recent years, rapid progress has been made in the application of ICIs in patients with advanced malignant tumors. IH has received increasing attention as a common endocrine irAE. On the one hand, the clinical manifestations of IH lack specificity. On the other hand, anterior pituitary insufficiency can threaten the safety of patients. Interrupting ICI therapy can lead to tumor progression. Therefore, early and correct diagnosis of IH is particularly important. The pathogenesis of IH caused by CTLA-4 inhibitors and PD-1/PD-L1 inhibitors is different. CTLA-4 inhibitor-induced IH is often accompanied by total anterior pituitary hypofunction combined with pituitary enlargement, while PD-1/PD-L1 inhibitor-induced IH is mainly characterized by isolated ACTH deficiency. Assessment of anterior pituitary hormones, especially the HPA axis, is one of the more sensitive methods for diagnosing IH. Dynamic monitoring of pituitary-target gland axis hormones and pituitary MRI is necessary before and during the use of ICIs. Once IH has been diagnosed, hormone replacement therapy should be started immediately, and high-dose glucocorticoid pulse therapy should be used with caution. It should be evaluated whether to suspend ICIs treatment based on the risk of benefit and harm. Timely and accurate diagnosis and treatment of IH has important clinical significance for ensuring patient safety and improving patient prognosis.

Author Contributions

P.C. and H.T. were the major contributors to writing and organizing documents. J.L. revised and reviewed the manuscript. All authors have read and agreed to the published version of the manuscript.

Funding

This work was supported by the Science and Technology Department of Sichuan Province (Grant No. 2018HH0065), the Grant 1.3.5 project for Disciplines of Excellence Clinical Research Incubation Project, West China Hospital Sichuan University (Grant No. 2020HXFH034) (J.L.) and the Ganbao Project of the Health Commission of Sichuan Province (Grant No. GBKT22014) (H.T.).

Data Availability Statement

Data Availability Statement:No new data were created.

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Zeng, H.-L.; Gao, X. Tumor immune checkpoint inhibitors-related endocrinopathies. Chin. J. Clin. Med. 2019, 26, 810–816. [Google Scholar]
  2. Wang, D.Y.; Salem, J.E.; Cohen, J.V.; Chandra, S.; Menzer, C.; Ye, F.; Zhao, S.; Das, S.; Beckermann, K.E.; Ha, L.; et al. Fatal Toxic Effects Associated With Immune Checkpoint Inhibitors: A Systematic Review and Meta-analysis. JAMA Oncol. 2018, 4, 1721–1728. [Google Scholar] [CrossRef]
  3. Kennedy, L.B.; Salama, A.K.S. A review of cancer immunotherapy toxicity. CA Cancer J. Clin. 2020, 70, 86–104. [Google Scholar] [CrossRef] [PubMed]
  4. Faje, A. Immunotherapy and hypophysitis: Clinical presentation, treatment, and biologic insights. Pituitary 2016, 19, 82–92. [Google Scholar] [CrossRef] [PubMed]
  5. Schnell, A.; Bod, L.; Madi, A.; Kuchroo, V.K. The yin and yang of co-inhibitory receptors: Toward anti-tumor immunity without autoimmunity. Cell Res. 2020, 30, 285–299. [Google Scholar] [CrossRef] [PubMed]
  6. Abril-Rodriguez, G.; Ribas, A. SnapShot: Immune Checkpoint Inhibitors. Cancer Cell 2017, 31, 848–848.e1. [Google Scholar] [CrossRef] [PubMed]
  7. Helmy, K.Y.; Patel, S.A.; Nahas, G.R.; Rameshwar, P. Cancer immunotherapy: Accomplishments to date and future promise. Ther. Deliv. 2013, 4, 1307–1320. [Google Scholar] [CrossRef]
  8. Puzanov, I.; Diab, A.; Abdallah, K.; Bingham, C.O., 3rd; Brogdon, C.; Dadu, R.; Hamad, L.; Kim, S.; Lacouture, M.E.; LeBoeuf, N.R.; et al. Managing toxicities associated with immune checkpoint inhibitors: Consensus recommendations from the Society for Immunotherapy of Cancer (SITC) Toxicity Management Working Group. J. Immunother. Cancer 2017, 5, 95. [Google Scholar] [CrossRef]
  9. Joshi, M.N.; Whitelaw, B.C.; Palomar, M.T.; Wu, Y.; Carroll, P.V. Immune checkpoint inhibitor-related hypophysitis and endocrine dysfunction: Clinical review. Clin. Endocrinol. 2016, 85, 331–339. [Google Scholar] [CrossRef]
  10. Haanen, J.; Carbonnel, F.; Robert, C.; Kerr, K.M.; Peters, S.; Larkin, J.; Jordan, K.; Committee, E.G. Management of toxicities from immunotherapy: ESMO Clinical Practice Guidelines for diagnosis, treatment and follow-up. Ann. Oncol. 2017, 28, iv119–iv142. [Google Scholar] [CrossRef]
  11. Brahmer, J.R.; Lacchetti, C.; Schneider, B.J.; Atkins, M.B.; Brassil, K.J.; Caterino, J.M.; Chau, I.; Ernstoff, M.S.; Gardner, J.M.; Ginex, P.; et al. Management of Immune-Related Adverse Events in Patients Treated With Immune Checkpoint Inhibitor Therapy: American Society of Clinical Oncology Clinical Practice Guideline. J. Clin. Oncol. 2018, 36, 1714–1768. [Google Scholar] [CrossRef] [PubMed]
  12. Higham, C.E.; Olsson-Brown, A.; Carroll, P.; Cooksley, T.; Larkin, J.; Lorigan, P.; Morganstein, D.; Trainer, P.J. Society for Endocrinology Endocrine Emergency Guidance: Acute management of the endocrine complications of checkpoint inhibitor therapy. Endocr. Connect. 2018, 7, G1–G7. [Google Scholar] [CrossRef] [PubMed]
  13. Arima, H.; Iwama, S.; Inaba, H.; Ariyasu, H.; Makita, N.; Otsuki, M.; Kageyama, K.; Imagawa, A.; Akamizu, T. Management of immune-related adverse events in endocrine organs induced by immune checkpoint inhibitors: Clinical guidelines of the Japan Endocrine Society. Endocr. J. 2019, 66, 581–586. [Google Scholar] [CrossRef] [PubMed]
  14. Castinetti, F.; Albarel, F.; Archambeaud, F.; Bertherat, J.; Bouillet, B.; Buffier, P.; Briet, C.; Cariou, B.; Caron, P.; Chabre, O.; et al. French Endocrine Society Guidance on endocrine side effects of immunotherapy. Endocr. Relat. Cancer 2019, 26, G1–G18. [Google Scholar] [CrossRef]
  15. Immunoendocrinology Group of Endocrinology Branch of Chinese Medical Association. Chinese expert consensus on the immune checkpoint inhibitors-induced endocrine immune-related adverse events (2020). Chin. J. Endocrinol. Metab. 2021, 37, 1–16. [Google Scholar]
  16. Wu, Y.Z.; Wu, Q.N.; Pu, D.L.; Zou, D.L.; Chen, B.; Wang, D.B.; Cai, J.L.; Wang, S.S.; Xiang, Y.; Yang, G.Y.; et al. Chinese expert consensus on immune checkpoint inhibitors induced emergency management of endocrine adverse reactions. J. Chongqing Med. Univ. 2021, 24, 1–12. [Google Scholar] [CrossRef]
  17. Corsello, S.M.; Barnabei, A.; Marchetti, P.; De Vecchis, L.; Salvatori, R.; Torino, F. Endocrine side effects induced by immune checkpoint inhibitors. J. Clin. Endocrinol. Metab. 2013, 98, 1361–1375. [Google Scholar] [CrossRef]
  18. Barroso-Sousa, R.; Barry, W.T.; Garrido-Castro, A.C.; Hodi, F.S.; Min, L.; Krop, I.E.; Tolaney, S.M. Incidence of Endocrine Dysfunction Following the Use of Different Immune Checkpoint Inhibitor Regimens: A Systematic Review and Meta-analysis. JAMA Oncol. 2018, 4, 173–182. [Google Scholar] [CrossRef]
  19. Zhang, B.; Wu, Q.; Zhou, Y.L.; Guo, X.; Ge, J.; Fu, J. Immune-related adverse events from combination immunotherapy in cancer patients: A comprehensive meta-analysis of randomized controlled trials. Int. Immunopharmacol. 2018, 63, 292–298. [Google Scholar] [CrossRef]
  20. Lu, J.; Li, L.; Lan, Y.; Liang, Y.; Meng, H. Immune checkpoint inhibitor-associated pituitary-adrenal dysfunction: A systematic review and meta-analysis. Cancer Med. 2019, 8, 7503–7515. [Google Scholar] [CrossRef]
  21. Phan, G.Q.; Yang, J.C.; Sherry, R.M.; Hwu, P.; Topalian, S.L.; Schwartzentruber, D.J.; Restifo, N.P.; Haworth, L.R.; Seipp, C.A.; Freezer, L.J.; et al. Cancer regression and autoimmunity induced by cytotoxic T lymphocyte-associated antigen 4 blockade in patients with metastatic melanoma. Proc. Natl. Acad. Sci. USA 2003, 100, 8372–8377. [Google Scholar] [CrossRef]
  22. Ishikawa, M.; Oashi, K. Case of hypophysitis caused by nivolumab. J. Dermatol. 2017, 44, 109–110. [Google Scholar] [CrossRef] [PubMed]
  23. Byun, D.J.; Wolchok, J.D.; Rosenberg, L.M.; Girotra, M. Cancer immunotherapy—Immune checkpoint blockade and associated endocrinopathies. Nat. Rev. Endocrinol. 2017, 13, 195–207. [Google Scholar] [CrossRef] [PubMed]
  24. Chang, L.S.; Yialamas, M.A. Checkpoint Inhibitor-Associated Hypophysitis. J. Gen. Intern. Med. 2018, 33, 125–127. [Google Scholar] [CrossRef]
  25. Yue, Y.; Jiang, T. Overview of Endocrine Toxicity Associated with Immunosuppressive Checkpoint Inhibitors. World Latest Med. Inf. 2020, 20, 134–136. [Google Scholar]
  26. El Osta, B.; Hu, F.; Sadek, R.; Chintalapally, R.; Tang, S.C. Not all immune-checkpoint inhibitors are created equal: Meta-analysis and systematic review of immune-related adverse events in cancer trials. Crit. Rev. Oncol. Hematol. 2017, 119, 1–12. [Google Scholar] [CrossRef]
  27. Wang, P.F.; Chen, Y.; Song, S.Y.; Wang, T.J.; Ji, W.J.; Li, S.W.; Liu, N.; Yan, C.X. Immune-Related Adverse Events Associated with Anti-PD-1/PD-L1 Treatment for Malignancies: A Meta-Analysis. Front. Pharmacol. 2017, 8, 730. [Google Scholar] [CrossRef]
  28. Baxi, S.; Yang, A.; Gennarelli, R.L.; Khan, N.; Wang, Z.; Boyce, L.; Korenstein, D. Immune-related adverse events for anti-PD-1 and anti-PD-L1 drugs: Systematic review and meta-analysis. BMJ 2018, 360, k793. [Google Scholar] [CrossRef]
  29. de Filette, J.; Andreescu, C.E.; Cools, F.; Bravenboer, B.; Velkeniers, B. A Systematic Review and Meta-Analysis of Endocrine-Related Adverse Events Associated with Immune Checkpoint Inhibitors. Horm. Metab. Res. 2019, 51, 145–156. [Google Scholar] [CrossRef]
  30. Xu, H.; Tan, P.; Zheng, X.; Huang, Y.; Lin, T.; Wei, Q.; Ai, J.; Yang, L. Immune-related adverse events following administration of anti-cytotoxic T-lymphocyte-associated protein-4 drugs: A comprehensive systematic review and meta-analysis. Drug Des. Dev. Ther. 2019, 13, 2215–2234. [Google Scholar] [CrossRef]
  31. Almutairi, A.R.; McBride, A.; Slack, M.; Erstad, B.L.; Abraham, I. Potential Immune-Related Adverse Events Associated With Monotherapy and Combination Therapy of Ipilimumab, Nivolumab, and Pembrolizumab for Advanced Melanoma: A Systematic Review and Meta-Analysis. Front. Oncol. 2020, 10, 91. [Google Scholar] [CrossRef] [PubMed]
  32. Xing, P.; Zhang, F.; Wang, G.; Xu, Y.; Li, C.; Wang, S.; Guo, Y.; Cai, S.; Wang, Y.; Li, J. Incidence rates of immune-related adverse events and their correlation with response in advanced solid tumours treated with NIVO or NIVO + IPI: A systematic review and meta-analysis. J. Immunother. Cancer 2019, 7, 341. [Google Scholar] [CrossRef] [PubMed]
  33. Da, L.; Teng, Y.; Wang, N.; Zaguirre, K.; Liu, Y.; Qi, Y.; Song, F. Organ-Specific Immune-Related Adverse Events Associated With Immune Checkpoint Inhibitor Monotherapy Versus Combination Therapy in Cancer: A Meta-Analysis of Randomized Controlled Trials. Front. Pharmacol. 2019, 10, 1671. [Google Scholar] [CrossRef] [PubMed]
  34. Ascierto, P.A.; Del Vecchio, M.; Robert, C.; Mackiewicz, A.; Chiarion-Sileni, V.; Arance, A.; Lebbé, C.; Bastholt, L.; Hamid, O.; Rutkowski, P.; et al. Ipilimumab 10 mg/kg versus ipilimumab 3 mg/kg in patients with unresectable or metastatic melanoma: A randomised, double-blind, multicentre, phase 3 trial. Lancet Oncol. 2017, 18, 611–622. [Google Scholar] [CrossRef]
  35. Jing, Y.; Zhang, Y.; Wang, J.; Li, K.; Chen, X.; Heng, J.; Gao, Q.; Ye, Y.; Zhang, Z.; Liu, Y.; et al. Association Between Sex and Immune-Related Adverse Events During Immune Checkpoint Inhibitor Therapy. J. Natl. Cancer Inst. 2021, 113, 1396–1404. [Google Scholar] [CrossRef] [PubMed]
  36. Faje, A.T.; Sullivan, R.; Lawrence, D.; Tritos, N.A.; Fadden, R.; Klibanski, A.; Nachtigall, L. Ipilimumab-induced hypophysitis: A detailed longitudinal analysis in a large cohort of patients with metastatic melanoma. J. Clin. Endocrinol. Metab. 2014, 99, 4078–4085. [Google Scholar] [CrossRef]
  37. Levy, M.; Abeillon, J.; Dalle, S.; Assaad, S.; Borson-Chazot, F.; Disse, E.; Raverot, G.; Cugnet-Anceau, C. Anti-PD1 and Anti-PDL1-Induced Hypophysitis: A Cohort Study of 17 Patients with Longitudinal Follow-Up. J. Clin. Med. 2020, 9, 3280. [Google Scholar] [CrossRef] [PubMed]
  38. Faje, A.; Reynolds, K.; Zubiri, L.; Lawrence, D.; Cohen, J.V.; Sullivan, R.J.; Nachtigall, L.; Tritos, N. Hypophysitis secondary to nivolumab and pembrolizumab is a clinical entity distinct from ipilimumab-associated hypophysitis. Eur. J. Endocrinol. 2019, 181, 211–219. [Google Scholar] [CrossRef]
  39. Briet, C.; Albarel, F.; Kuhn, E.; Merlen, E.; Chanson, P.; Cortet, C. Expert opinion on pituitary complications in immunotherapy. Ann. Endocrinol. 2018, 79, 562–568. [Google Scholar] [CrossRef]
  40. Girotra, M.; Hansen, A.; Farooki, A.; Byun, D.J.; Min, L.; Creelan, B.C.; Callahan, M.K.; Atkins, M.B.; Sharon, E.; Antonia, S.J.; et al. The Current Understanding of the Endocrine Effects From Immune Checkpoint Inhibitors and Recommendations for Management. JNCI Cancer Spectr. 2018, 2, pky021. [Google Scholar] [CrossRef]
  41. Boudjemaa, A.; Rousseau-Bussac, G.; Monnet, I. Late-Onset Adrenal Insufficiency More Than 1 Year after Stopping Pembrolizumab. J. Thorac. Oncol. 2018, 13, e39–e40. [Google Scholar] [CrossRef]
  42. Iwama, S.; De Remigis, A.; Callahan, M.K.; Slovin, S.F.; Wolchok, J.D.; Caturegli, P. Pituitary expression of CTLA-4 mediates hypophysitis secondary to administration of CTLA-4 blocking antibody. Sci. Transl. Med. 2014, 6, 230ra245. [Google Scholar] [CrossRef]
  43. Caturegli, P.; Di Dalmazi, G.; Lombardi, M.; Grosso, F.; Larman, H.B.; Larman, T.; Taverna, G.; Cosottini, M.; Lupi, I. Hypophysitis Secondary to Cytotoxic T-Lymphocyte-Associated Protein 4 Blockade: Insights into Pathogenesis from an Autopsy Series. Am. J. Pathol. 2016, 186, 3225–3235. [Google Scholar] [CrossRef]
  44. Di Dalmazi, G.; Ippolito, S.; Lupi, I.; Caturegli, P. Hypophysitis induced by immune checkpoint inhibitors: A 10-year assessment. Expert Rev. Endocrinol. Metab. 2019, 14, 381–398. [Google Scholar] [CrossRef]
  45. Vidarsson, G.; Dekkers, G.; Rispens, T. IgG subclasses and allotypes: From structure to effector functions. Front. Immunol. 2014, 5, 520. [Google Scholar] [CrossRef]
  46. Chang, L.S.; Barroso-Sousa, R.; Tolaney, S.M.; Hodi, F.S.; Kaiser, U.B.; Min, L. Endocrine Toxicity of Cancer Immunotherapy Targeting Immune Checkpoints. Endocr. Rev. 2019, 40, 17–65. [Google Scholar] [CrossRef]
  47. Li, J.Y.; Xing, B. Research progress of immune checkpoint inhibitor related autoimmune hypophysitis. Basic Clin. Med. 2020, 40, 403–406. [Google Scholar]
  48. Amirbaigloo, A.; Esfahanian, F.; Mouodi, M.; Rakhshani, N.; Zeinalizadeh, M. IgG4-related hypophysitis. Endocrine 2021, 73, 270–291. [Google Scholar] [CrossRef]
  49. Landek-Salgado, M.A.; Leporati, P.; Lupi, I.; Geis, A.; Caturegli, P. Growth hormone and proopiomelanocortin are targeted by autoantibodies in a patient with biopsy-proven IgG4-related hypophysitis. Pituitary 2012, 15, 412–419. [Google Scholar] [CrossRef]
  50. Iwata, N.; Iwama, S.; Sugimura, Y.; Yasuda, Y.; Nakashima, K.; Takeuchi, S.; Hagiwara, D.; Ito, Y.; Suga, H.; Goto, M.; et al. Anti-pituitary antibodies against corticotrophs in IgG4-related hypophysitis. Pituitary 2017, 20, 301–310. [Google Scholar] [CrossRef]
  51. Kanie, K.; Iguchi, G.; Bando, H.; Urai, S.; Shichi, H.; Fujita, Y.; Matsumoto, R.; Suda, K.; Yamamoto, M.; Fukuoka, H.; et al. Mechanistic insights into immune checkpoint inhibitor-related hypophysitis: A form of paraneoplastic syndrome. Cancer Immunol. Immunother. 2021, 70, 3669–3677. [Google Scholar] [CrossRef] [PubMed]
  52. Tahir, S.A.; Gao, J.; Miura, Y.; Blando, J.; Tidwell, R.S.S.; Zhao, H.; Subudhi, S.K.; Tawbi, H.; Keung, E.; Wargo, J.; et al. Autoimmune antibodies correlate with immune checkpoint therapy-induced toxicities. Proc. Natl. Acad. Sci. USA 2019, 116, 22246–22251. [Google Scholar] [CrossRef] [PubMed]
  53. Régnauld, K.L.; Leteurtre, E.; Gutkind, S.J.; Gespach, C.P.; Emami, S. Activation of adenylyl cyclases, regulation of insulin status, and cell survival by G(alpha)olf in pancreatic beta-cells. Am. J. Physiol. Regul. Integr. Comp. Physiol. 2002, 282, R870–R880. [Google Scholar] [CrossRef]
  54. Audo, I.; Bujakowska, K.; Orhan, E.; El Shamieh, S.; Sennlaub, F.; Guillonneau, X.; Antonio, A.; Michiels, C.; Lancelot, M.E.; Letexier, M.; et al. The familial dementia gene revisited: A missense mutation revealed by whole-exome sequencing identifies ITM2B as a candidate gene underlying a novel autosomal dominant retinal dystrophy in a large family. Hum. Mol. Genet. 2014, 23, 491–501. [Google Scholar] [CrossRef]
  55. Iglesias, P.; Sánchez, J.C.; Díez, J.J. Isolated ACTH deficiency induced by cancer immunotherapy: A systematic review. Pituitary 2021, 24, 630–643. [Google Scholar] [CrossRef] [PubMed]
  56. Ihara, K. Immune checkpoint inhibitor therapy for pediatric cancers: A mini review of endocrine adverse events. Clin. Pediatr. Endocrinol. 2019, 28, 59–68. [Google Scholar] [CrossRef]
  57. Lupu, J.; Pages, C.; Laly, P.; Delyon, J.; Laloi, M.; Petit, A.; Basset-Seguin, N.; Oueslati, I.; Zagdanski, A.M.; Young, J.; et al. Transient pituitary ACTH-dependent Cushing syndrome caused by an immune checkpoint inhibitor combination. Melanoma Res. 2017, 27, 649–652. [Google Scholar] [CrossRef]
  58. Dillard, T.; Yedinak, C.G.; Alumkal, J.; Fleseriu, M. Anti-CTLA-4 antibody therapy associated autoimmune hypophysitis: Serious immune related adverse events across a spectrum of cancer subtypes. Pituitary 2010, 13, 29–38. [Google Scholar] [CrossRef]
  59. Nallapaneni, N.N.; Mourya, R.; Bhatt, V.R.; Malhotra, S.; Ganti, A.K.; Tendulkar, K.K. Ipilimumab-induced hypophysitis and uveitis in a patient with metastatic melanoma and a history of ipilimumab-induced skin rash. J. Natl. Compr. Cancer Netw. 2014, 12, 1077–1081. [Google Scholar] [CrossRef]
  60. Deligiorgi, M.V.; Siasos, G.; Vergadis, C.; Trafalis, D.T. Central diabetes insipidus related to anti-programmed cell-death 1 protein active immunotherapy. Int. Immunopharmacol. 2020, 83, 106427. [Google Scholar] [CrossRef]
  61. Fosci, M.; Pigliaru, F.; Salcuni, A.S.; Ghiani, M.; Cherchi, M.V.; Calia, M.A.; Loviselli, A.; Velluzzi, F. Diabetes insipidus secondary to nivolumab-induced neurohypophysitis and pituitary metastasis. Endocrinol. Diabetes Metab. Case Rep. 2021, 2021, 20–0123. [Google Scholar] [CrossRef] [PubMed]
  62. Zhao, C.; Tella, S.H.; Del Rivero, J.; Kommalapati, A.; Ebenuwa, I.; Gulley, J.; Strauss, J.; Brownell, I. Anti-PD-L1 Treatment Induced Central Diabetes Insipidus. J. Clin. Endocrinol. Metab. 2018, 103, 365–369. [Google Scholar] [CrossRef] [PubMed]
  63. Albarel, F.; Castinetti, F.; Brue, T. Management of Endocrine Disease: Immune check point inhibitors-induced hypophysitis. Eur. J. Endocrinol. 2019, 181, R107–R118. [Google Scholar] [CrossRef]
  64. Castinetti, F.; Albarel, F.; Archambeaud, F.; Bertherat, J.; Bouillet, B.; Buffier, P.; Briet, C.; Cariou, B.; Caron, P.; Chabre, O.; et al. Endocrine side-effects of new anticancer therapies: Overall monitoring and conclusions. Ann. Endocrinol. 2018, 79, 591–595. [Google Scholar] [CrossRef]
  65. Haanen, J.; Obeid, M.; Spain, L.; Carbonnel, F.; Wang, Y.; Robert, C.; Lyon, A.R.; Wick, W.; Kostine, M.; Peters, S.; et al. Management of toxicities from immunotherapy: ESMO Clinical Practice Guideline for diagnosis, treatment and follow-up. Ann. Oncol. 2022, 33, 1217–1238. [Google Scholar] [CrossRef]
  66. Min, L.; Hodi, F.S.; Giobbie-Hurder, A.; Ott, P.A.; Luke, J.J.; Donahue, H.; Davis, M.; Carroll, R.S.; Kaiser, U.B. Systemic high-dose corticosteroid treatment does not improve the outcome of ipilimumab-related hypophysitis: A retrospective cohort study. Clin. Cancer Res. 2015, 21, 749–755. [Google Scholar] [CrossRef] [PubMed]
  67. Iglesias, P. Cancer immunotherapy-induced endocrinopathies: Clinical behavior and therapeutic approach. Eur. J. Intern. Med. 2018, 47, 6–13. [Google Scholar] [CrossRef] [PubMed]
  68. Faje, A.T.; Lawrence, D.; Flaherty, K.; Freedman, C.; Fadden, R.; Rubin, K.; Cohen, J.; Sullivan, R.J. High-dose glucocorticoids for the treatment of ipilimumab-induced hypophysitis is associated with reduced survival in patients with melanoma. Cancer 2018, 124, 3706–3714. [Google Scholar] [CrossRef]
  69. Albarel, F.; Gaudy, C.; Castinetti, F.; Carré, T.; Morange, I.; Conte-Devolx, B.; Grob, J.J.; Brue, T. Long-term follow-up of ipilimumab-induced hypophysitis, a common adverse event of the anti-CTLA-4 antibody in melanoma. Eur. J. Endocrinol. 2015, 172, 195–204. [Google Scholar] [CrossRef]
Figure 1. The diagnosis, treatment and follow-up process of ICI-induced hypophysitis. Note: a: laboratory tests; ACTH, cortisol (8:00), TSH, FT4, FSH, LH and T (men) or FSH, LH and E (premenopausal women) or FSH (postmenopausal women); PRL, IGF-1.; blood sodium, blood and urine osmotic pressure.
Figure 1. The diagnosis, treatment and follow-up process of ICI-induced hypophysitis. Note: a: laboratory tests; ACTH, cortisol (8:00), TSH, FT4, FSH, LH and T (men) or FSH, LH and E (premenopausal women) or FSH (postmenopausal women); PRL, IGF-1.; blood sodium, blood and urine osmotic pressure.
Jcm 12 03468 g001
Table 1. Representative drugs and indications of CTLA-4 inhibitors and PD-1/PD-L1 inhibitors.
Table 1. Representative drugs and indications of CTLA-4 inhibitors and PD-1/PD-L1 inhibitors.
Drug InformationCTLA-4 InhibitorsPD-1 InhibitorsPD-L1 Inhibitors
IpilimumabTremelimumabNivolumabPembrolizumabCemiplimabToripalimabSintilimabCamrelizumabTislelizumabAtezolizumabAvelumabDurvalumab
Drug nameYervoyNo brandOpdivoKeytrudaLibtayoTuoyiDaboshuEricaBaizeanTecentriqBavencioImfinzi
IgG typeIgG1IgG2IgG4IgG4IgG4IgG4IgG4IgG4kIgG4IgG1kIgG1IgG1k
Main indicationmelanomaHepatocellular cancer, malignant mesotheliomaMelanoma, Esophageal squamous cell cancer and other tumors aMelanoma, metastatic NSCLC and other tumors bCSCCMelanoma, nasopharyngeal cancerClassical Hodgkin’s lymphomaClassical Hodgkin’s lymphomaClassical Hodgkin’s lymphoma, urothelial cancerUrothelial cancer, NSCLCMerkel cell cancer, urothelial cancerUrothelial cancer, NSCLC
Number of ongoing trials4951211088142580291264301244557167473
Note: NSCLC, non-small-cell lung cancer; CSCC, squamous cell carcinoma of the skin. a: NSCLC; CSCC; renal cell cancer; small cell lung cancer; classical Hodgkin’s lymphoma; urothelial cancer; head and neck Squamous cancer; microsatellite instability–high/deficient mismatch repair cancer; colorectal cancer; hepatocellular cancer. b: classical Hodgkin’s lymphoma; large B-cell lymphoma; Merkel cell cancer; head and neck squamous cell cancer; urothelial cancer; microsatellite instability–high/deficient mismatch repair cancer; gastric cancer; cervical cancer; hepatocellular cancer.
Table 2. Incidence of ICI-induced hypophysitis reported in the literature from 2017 to the present.
Table 2. Incidence of ICI-induced hypophysitis reported in the literature from 2017 to the present.
AuthorCTLA-4 Inhibitors Combination PD-1/PD-L1 InhibitorsCTLA-4 InhibitorsPD-1/PD-L1 Inhibitors
CTCAE1 to 5CTCAE3 to 5CTCAE1 to 5C TCAE3 to 5CTCAE1 to 5CTCAE3 to 5
Barroso-Sousa et al. [18]6.4% (Ipi + Niv)NA3.2%NA0.5%NA
El Osta et al. [26]8.0%1.7%5.40%3.3%0.30%0.1%
Wang et al. [27]NANANANA0.85%0.59%
Baxi et al. [28].NANANANA0.3%0.2%
De Filette et al. [29].8.8% (Ipi + Niv), 10.5% (Ipi + Pem)NA1.8%(Tre), 5.6% (Ipi)NA0.5% (Niv), 1.1%(Pem)NA
Xu et al. [30].NANA3.3%1.7%NANA
Wang et al. [2]NA2.0%NA5.0%NA1%
Zhang et al. [19]9.7% NANANANA
Almutairi et al. [31]10.4% (Ipi + Niv), 10.46% (Ipi + Pem)2.48% (Ipi + Niv)4.13% (Ipi)1.21% (Ipi)0.31% (Niv), 0.66%(Pem)0.15% (Niv), 0.68% (Pem)
Lu et al. [20]7.68%1.66%4.53%0.78%0.41%0.06%
Xing et al. [32]8.38% (Ipi + Niv)2.48% (Ipi + Niv)NANA0.47% (Niv)0.35% (Niv)
Li et al. [33] 10%1.1%4.2%1.7%0.6%1.5%
Note: CTCAE, the common terminology criteria for adverse events; Ipi, ipilimumab; Niv, nivolumab; Pem, pembrolizumab; Tre, tremelimumab; Atz, atezolizumab; Ave, avelumab; NA, not available.
Table 3. Differences between the two types of IH.
Table 3. Differences between the two types of IH.
Clinical ManifestationsCTLA-4 Inhibitors-Induced HypophysitisPD-1/PD-L1 Inhibitors-Induced Hypophysitis
The median onset age60 years60~65 years
Gender differenceMale: female 4:1Male: female 1:1~3:1
The median onset time9.3 weeks28 weeks
Clinical manifestationsHeadache and fatigue as the first manifestation, accompanied by visual impairment, hypothyroidism, loss of libido (hypohypophysis).Fatigue and loss of appetite as the first manifestation, accompanied by weight loss, hypotension, etc. (isolated ACTH deficiency)
Laboratory testsACTH deficiency 55%; TSH deficiency 81%; FSH/LH deficiency 88%; Hyponatremia 45%; 12.5% lower IGF—112.5%;ACTH deficiency of 100%; TSH deficiency 11.8%; FSH/LH deficiency 18.8%; Hyponatremia 52.9%; lower IGF—113.3%;
Pituitary MRIpituitary enlargement, stalk enlargement, or enhancement changes after injection of contrast agent often occur, an incidence of 98%pituitary enlargement rate 28%
Note: ACTH, adrenocorticotropic hormone; TSH, thyroid stimulating hormone; FSH, follicle-stimulating hormone; LH, luteinizing hormone; IGF-1, insulin-like growth factor 1.
Table 4. CTCAE grading and specific clinical manifestations of ICI-induced hypophysitis.
Table 4. CTCAE grading and specific clinical manifestations of ICI-induced hypophysitis.
GradeDescription of CTCAEClinical Manifestations
Level 1Mild: Asymptomatic or mildly symptomatic.Mild fatigue or anorexia, without headache or asymptomatic.
Level 2Moderate: Mild-to-moderate, mild local symptoms; noninvasive treatment needed; mild impairment of self-care; no limitation of daily activities.Headache without visual disturbances or fatigue, mood changes without hemodynamic instability and electrolyte disturbances.
Level 3Severe: Severe or clinically important but not temporarily life-threatening; requiring hospitalization or prolonged hospitalization; severely impaired self-care ability; affecting daily activities.Headache with visual disturbances, or adrenal insufficiency with hypotension, electrolyte disturbances.
Level 4Life-Threatening: Life-threatening, requiring urgent intervention; inability to perform daily activities.Severe headache with visual disturbance, or severe adrenal insufficiency with shock, severe electrolyte.
Level 5Death.Death.
Note: CTCAE, the common terminology criteria for adverse events.
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.

Share and Cite

MDPI and ACS Style

Chen, P.; Li, J.; Tan, H. Progress and Challenges of Immune Checkpoint Inhibitor-Induced Hypophysitis. J. Clin. Med. 2023, 12, 3468. https://doi.org/10.3390/jcm12103468

AMA Style

Chen P, Li J, Tan H. Progress and Challenges of Immune Checkpoint Inhibitor-Induced Hypophysitis. Journal of Clinical Medicine. 2023; 12(10):3468. https://doi.org/10.3390/jcm12103468

Chicago/Turabian Style

Chen, Piaohong, Jianwei Li, and Huiwen Tan. 2023. "Progress and Challenges of Immune Checkpoint Inhibitor-Induced Hypophysitis" Journal of Clinical Medicine 12, no. 10: 3468. https://doi.org/10.3390/jcm12103468

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop