ﺑﺎﺯﮔﺸﺖ ﺑﻪ ﺻﻔﺤﻪ ﻗﺒﻠﯽ
خرید پکیج
تعداد آیتم قابل مشاهده باقیمانده : 3 مورد
نسخه الکترونیک
medimedia.ir

Toxicities associated with immune checkpoint inhibitors

Toxicities associated with immune checkpoint inhibitors
Literature review current through: Jan 2024.
This topic last updated: Jan 03, 2024.

INTRODUCTION — Immune checkpoint inhibitors (ICIs), also known as checkpoint inhibitor immunotherapy, are immunomodulatory antibodies that are used to enhance the immune system. These agents have substantially improved the prognosis for patients with many advanced malignancies.

The primary targets for checkpoint inhibition include (table 1):

Programmed cell death receptor 1 and programmed cell death ligand 1 – Multiple antibodies against programmed cell death receptor 1 (PD-1) and programmed cell death ligand 1 (PD-L1) have been approved by the US Food and Drug Administration (FDA) or are in development and have shown great promise in multiple malignancies. Nivolumab, pembrolizumab, cemiplimab, dostarlimab, retifanlimab, and toripalimab, all of which target PD-1, and atezolizumab, avelumab, and durvalumab, all of which target PD-L1, have been approved for various indications.

Cytotoxic T lymphocyte-associated antigen 4Ipilimumab and tremelimumab, anti-cytotoxic T lymphocyte-associated antigen 4 (CTLA-4) antibodies, are approved by the FDA, with others in development.

Despite important clinical benefits, ICIs are associated with a unique spectrum of side effects known as immune-related adverse events (irAEs) [1,2]. IrAEs include dermatologic, gastrointestinal, hepatic, endocrine, and other less common inflammatory events. IrAEs are believed to arise from general immunologic enhancement, and temporary immunosuppression with glucocorticoids, tumor necrosis factor-alpha antagonists, mycophenolate mofetil, or other agents can be an effective treatment in most cases. Although rare, fulminant and even fatal toxicities may occur with ICIs [3,4]. Therefore, prompt recognition and management of irAEs is important.

The side effects of the checkpoint-blocking antibodies targeting the PD-1 and PD-L1 receptors and CTLA-4 are reviewed here. The management approach to irAEs is presented based on clinical experience, since no prospective trials have been conducted to guide the treatment of irAEs. Principles of cancer immunotherapy are presented separately. (See "Principles of cancer immunotherapy".)

GENERAL PRINCIPLES — The American Society of Clinical Oncology (ASCO) organized a multidisciplinary panel that reviewed the literature and proposed both general guidelines and organ-systems-specific recommendations for the management of adverse events associated with ICIs [5]. The general approach is outlined in this section, and more specific recommendations are discussed below for the various toxicities. Our approach is consistent with those recommendations and those of the Society for Immunotherapy of Cancer [6].

Dose modifications and immunosuppressive therapy — In general, treatment of moderate or severe immune-related adverse events (irAEs) requires interruption of ICIs and immunosuppression with glucocorticoids. Patients should be carefully monitored during treatment for initial evidence of grade 1 adverse events. Treatment is based on the site of the irAE and the severity of the observed toxicity:

Grade 1 irAEs – For most patients with grade 1 irAEs (asymptomatic or mild symptoms), immunotherapy may be continued, and patients may be closely monitored for worsening irAEs and/or managed symptomatically.

Grade 2 irAEs – For most patients with grade 2 (moderate) immune-mediated toxicities (excluding endocrinopathies), treatment with immunotherapy should be withheld and should not be resumed until symptoms or toxicity is grade 1 or less. Glucocorticoids (prednisone 0.5 mg/kg/day or equivalent) should be started if symptoms do not resolve within one week.

Grade 2 endocrinopathies – An exception is for patients who experience immune-mediated endocrinopathies but do not require immunosuppressive therapy. In such patients, depending upon the severity of symptoms, immunotherapy may be withheld until hormone replacement is initiated. Immunotherapy may subsequently be resumed once acute symptoms have resolved and (in the event of adrenal insufficiency or hypophysitis) patients are receiving adequate adrenal glucocorticoid replacement. (See 'Endocrinopathies' below and 'Retreatment after prior toxicity' below.)

Grade 3 or 4 irAEs For patients experiencing grade 3 or 4 (severe or life-threatening) immune-mediated toxicities, treatment with immunotherapy is usually permanently discontinued. High doses of glucocorticoids (prednisone 1 to 2 mg/kg/day or equivalent) should be given. When symptoms subside to grade 1 or less, glucocorticoids can be gradually tapered, typically over at least four weeks (table 2).

Refractory toxicity – In the authors' experience, patients who will benefit from glucocorticoids generally do so within days to a week. If symptoms do not clearly improve or worsen on steroids, our approach is to administer an additional immunosuppressive agent. Examples of available agents include infliximab, vedolizumab, and mycophenolate mofetil, among other options.

The management of specific toxicities is discussed in the individual sections below.

Impact of immunosuppressive agents on immunotherapy efficacy — For patients who have required glucocorticoids or other immunosuppressive therapy, most data suggest that efficacy of the ICI is not impacted [7-9]. However, some studies suggest that either early use of corticosteroids (within two months) or second-line immunosuppressive therapy (in addition to corticosteroids) is associated with worsened survival outcomes [10,11]. Additionally, for patients being evaluated for retreatment with immunotherapy after experiencing an irAE, the concurrent use of immunosuppressive therapy is associated with reduced efficacy of immunotherapy. (See 'Retreatment after prior toxicity' below.)

Relationship between immunotherapy toxicities and efficacy — Most data suggest that irAEs are associated with either improved efficacy of immunotherapy (such as favorable response rates and prolonged survival [7,12-28]) or similar efficacy, compared with those without irAEs [8,29-32]. Although most clinicians commonly associate irAEs with treatment-related toxicity, the presence of an irAE is also a sign that the immune system is sufficiently activated to hopefully additionally target the patient's cancer. Therefore, management of irAEs should entail the minimum amount of immunosuppression needed to control symptoms and, in certain cases, holding therapy without initiating immunosuppression (eg, select immune-mediated endocrinopathies; asymptomatic hepatitis or pancreatitis). (See 'Dose modifications and immunosuppressive therapy' above and "Hepatic, pancreatic, and rare gastrointestinal complications of immune checkpoint inhibitor therapy".)

Predictive biomarkers of immunotherapy toxicity — The optimal biomarker to predict the risk of irAEs and as an aid in the early identification of such complications remains to be defined. In retrospective observational studies, biomarkers associated with higher risk of developing irAEs include clinical characteristics (eg, female sex [33]), germline and somatic genetic features, specific microbiome compositions [34], and circulating biomarkers [35].

Combining or sequencing immunotherapy with other therapies — In a variety of tumors, ICIs are being either combined or sequenced with other systemic agents (eg, chemotherapy, targeted therapies) and other treatment modalities (eg, radiation therapy [RT], allogeneic stem cell transplant) [36]. Such combinations result in varied toxicity profiles [37]. Clinicians should assess and treat the toxicities associated with each individual agent and remain aware of the potential for development of novel toxicities.

Immunotherapy plus chemotherapy – Details on the efficacy and toxicity of immunotherapy plus chemotherapy in non-small cell lung cancer are discussed separately. (See "Initial management of advanced non-small cell lung cancer lacking a driver mutation", section on 'PD-L1 low (<50 percent) or unselected tumors'.)

Immunotherapy plus targeted therapy – For certain tumors, such as lung cancer, some combinations of immunotherapy with targeted therapies (either administered concurrently or close together) have been associated with serious and even potentially life-threatening toxicities. Accordingly, given the long half-life of ICIs, we do not combine immunotherapy with targeted therapies outside of a clinical trial, unless the safety of the combination has already been demonstrated.

The combination of immunotherapy with agents targeting the epidermal growth factor receptor (EGFR) and anaplastic lymphoma kinase (ALK) pathways have been associated with life-threatening irAEs (eg, pneumonitis, hepatitis) in patients with advanced non-small cell lung cancer. This is discussed separately. (See "Systemic therapy for advanced non-small cell lung cancer with an activating mutation in the epidermal growth factor receptor", section on 'Immune-related toxicities with EGFR TKI after immunotherapy' and "Anaplastic lymphoma kinase (ALK) fusion oncogene positive non-small cell lung cancer", section on 'Efficacy of other approaches'.)

For other tumors, such as renal cell carcinoma, the combination of targeted therapy and immunotherapy has been more successful. Details on the efficacy and toxicity of immunotherapy plus antiangiogenic therapy targeting the vascular endothelial growth factor (VEGF) pathway in advanced renal cell carcinoma are discussed separately. (See "Systemic therapy of advanced clear cell renal carcinoma".)

In patients with metastatic melanoma, ICIs and BRAF plus MEK inhibitors have also been safely combined [38-43] and have regulatory approval in this setting. Further details on these combinations are discussed separately. (See "Systemic treatment of metastatic melanoma with BRAF and other molecular alterations", section on 'Is there a role for combined immunotherapy and targeted therapy?'.)

The combination of ipilimumab with BRAF and MEK inhibitors is not used in clinical practice because it has resulted in severe toxicities such as hepatotoxicity, rash, colitis, and intestinal perforation [38,39,44]. The individual efficacy of these agents in melanoma is discussed separately. (See "Systemic treatment of metastatic melanoma lacking a BRAF mutation" and "Systemic treatment of metastatic melanoma with BRAF and other molecular alterations".)

Radiation therapy followed by immunotherapy – Data suggest that immunotherapy can be safely used after completion of radiation therapy in most cancers. As an example, in one observational study of 16,835 patients with cancer treated with various ICI regimens, the sequential use of immunotherapy within 90 days following RT was not associated with significantly increased rates of irAEs, compared with those who did not receive RT prior to immunotherapy [45].

Allogeneic stem cell transplant after prior immunotherapy – The role of allogeneic stem cell transplantation after prior immunotherapy is discussed separately. (See "Treatment of extranodal NK/T cell lymphoma, nasal type", section on 'Relapsed or refractory disease' and "Treatment of relapsed or refractory classic Hodgkin lymphoma".)

SYSTEMIC ADVERSE EVENTS

Fatigue — Fatigue is among the most common side effects seen, with an estimated overall frequency of 16 to 24 percent for the anti-programmed cell death receptor 1 (PD-1) and anti-programmed cell death ligand 1 (PD-L1) agents [1] and approximately 26 percent in those treated with combination immunotherapy [46] However, the fatigue is generally mild, and severe fatigue is rare as a side effect of these agents. When fatigue is present, it is important to exclude thyroid, pituitary, and other endocrine disorders, such as primary adrenal insufficiency. Fever, chills, and infusion reactions have also been described, but these are also rare.

Infusion-related reactions — Mild infusion-related (grade 1 and 2) side effects have been reported in up to 25 percent of patients treated with agents that inhibit PD-1 or PD-L1. These infusion-related reactions are reported most commonly with avelumab, a PD-L1 inhibitor. For patients treated with avelumab, premedication with acetaminophen and an antihistamine is indicated during the first four cycles, and subsequently as needed [47]. We discourage premedicating or managing infusion-related reactions using glucocorticoids, because such reactions often improve with antihistamines alone. (See "Treatment of recurrent and metastatic Merkel cell carcinoma", section on 'Avelumab'.)

Immunotherapy is discontinued permanently for patients who experience life-threatening (grade 4) infusion reactions. The reported incidence of high-grade infusion-related reactions due to immunotherapy is rare (less than 2 percent). (See "Infusion-related reactions to therapeutic monoclonal antibodies used for cancer therapy", section on 'Agents targeting the programmed cell death receptor'.)

Management recommendations are summarized in the American Society of Clinical Oncology (ASCO) guidelines table (table 3) [5].

Cytokine release syndrome — Cytokine release syndrome (CRS) is an acute systemic inflammatory response syndrome characterized by fever with or without multiple organ dysfunction mostly described with chimeric antigen receptor (CAR)-T cell therapy. CRS has also been observed with ICIs, such as nivolumab [48,49]. The management of CRS in patients treated with ICIs and other antibody-mediated therapies is discussed separately. (See "Cytokine release syndrome (CRS)", section on 'Bi-specific antibody-associated CRS'.)

DERMATOLOGIC AND MUCOSAL TOXICITY — Cutaneous immune-related adverse events (irAEs) are the most common toxicity associated with ICIs. While the most common clinical manifestation is an inflammatory skin reaction, other presentations exist such as immunobullous disease, vasculitis, neutrophilic dermatoses, and, rarely, severe cutaneous drug reactions. The management of cutaneous irAEs is based upon severity and impact on the patient's functional status.

Further details on the clinical presentation, diagnosis, and management of cutaneous irAEs are discussed separately. (See "Cutaneous immune-related adverse events associated with immune checkpoint inhibitors".)

DIARRHEA/COLITIS — Diarrhea and colitis are the most frequently reported gastrointestinal immune-related adverse events (irAEs) in patients undergoing treatment with ICIs. Further details on the clinical presentation, diagnosis, and management of ICI colitis are discussed separately. (See "Immune checkpoint inhibitor colitis".)

HEPATOTOXICITY — Hepatotoxicity is a frequent complication of ICIs. Further details on the clinical presentation, diagnosis, and management of hepatotoxicity from ICIs are discussed separately. (See "Hepatic, pancreatic, and rare gastrointestinal complications of immune checkpoint inhibitor therapy".)

PNEUMONITIS

Epidemiology — Pneumonitis is an uncommon but potentially severe or fatal complication of treatment with ICI therapy [50-55], with an overall incidence of approximately 5 percent [52]. The incidence of ICI pneumonitis is lower in patients treated with an anti-programmed cell death receptor 1 (PD-1) or anti-programmed cell death ligand 1 (PD-L1) monoclonal antibody (3 percent) versus those treated with an ICI combination that includes an anti-CTLA-4 antibody (10 percent) [52]. ICI pneumonitis is also more common in patients with lung cancer [56,57].

Clinical manifestations — Patients with ICI pneumonitis most commonly present with symptoms of dyspnea and cough (53 and 35 percent, respectively), while approximately one-third of patients are asymptomatic [52]. Most patients with ICI pneumonitis present with grade 1 or grade 2 involvement (table 4). More than half of patients with ICI pneumonitis may also present with another immune-related adverse event (irAE), such as colitis, dermatitis, or thyroiditis. (See 'Diarrhea/colitis' above and 'Dermatologic and mucosal toxicity' above and 'Endocrinopathies' below.)

The timing between initiation of ICI therapy and development of ICI pneumonitis varies, with a median of 2.8 months (range 9 days to 19 months), and occurs earlier for those treated with combination ICI therapy rather than single-agent therapy (median 2.7 versus 4.6 months) [52]. In some circumstances, pneumonitis can also present as a delayed irAE and occur more than one year after initiating therapy [58]).

ICI pulmonary toxicity may also manifest as a radiation recall pneumonitis limited to previously irradiated areas of the lung [59]. This phenomenon can occur when treatment is initiated years after radiation therapy.

Diagnosis

When to suspect the diagnosis – The diagnosis of pneumonitis from ICI therapy should be suspected in patients on active treatment with an ICI who present with new or worsening cough, shortness of breath, dyspnea on exertion, and/or oxygen requirement. (See 'Clinical manifestations' above.)

Differential diagnosis – Drug-induced pneumonitis is a diagnosis of exclusion. Therefore, patients with suspected ICI pneumonitis should also be evaluated for alternative diagnoses such as pulmonary embolism, infection (including coronavirus disease 2019 [COVID-19], which can mimic ICI pneumonitis), malignant pulmonary infiltration, congestive heart failure, and chronic obstructive pulmonary disease (COPD) exacerbation in those with a history of smoking (table 5). (See "Epidemiology and pathogenesis of acute pulmonary embolism in adults" and "Overview of community-acquired pneumonia in adults" and "COVID-19: Considerations in patients with cancer" and "Clinical manifestations and diagnosis of advanced heart failure" and "COPD exacerbations: Clinical manifestations and evaluation".)

Diagnostic evaluation – For all patients with symptoms of ICI pneumonitis, the initial diagnostic evaluation includes physical examination and pulse oximetry. (See "Approach to the patient with dyspnea", section on 'Physical examination'.)

For patients with mild (grade 2 or lower (table 4)) symptoms, we obtain initial imaging with a chest radiograph (CXR). A CXR can be performed rapidly, and patients with normal imaging findings can often proceed with treatment if ICI pneumonitis is not suspected. In patients where there is a high index of suspicion for ICI pneumonitis (eg, persistent or progressive symptoms), we offer short-interval clinical follow-up and a contrast-enhanced computed tomography (CT) of the chest to reassess for mild or developing pneumonitis.

For patients with more severe (grade 3 or higher) symptoms or those with abnormal findings on an initial CXR, we refer to the emergency department for prompt evaluation, including a contrast-enhanced CT of the chest. A chest CT can demonstrate several radiographic patterns associated with ICI pneumonitis, although there is no one characteristic radiographic feature for this condition (figure 1). A contrast-enhanced chest CT can also assess for other potential diagnoses, such as pulmonary embolism, malignant pulmonary infiltration, or infection. In observational studies, a CXR did not detect new radiographic abnormalities in approximately one-fourth of patients with ICI pneumonitis [52,60-62].

Bronchoscopy is not usually necessary for the diagnosis of ICI pneumonitis; its use is typically reserved for patients with grade 2 or higher pneumonitis (table 4) whose diagnosis remains unclear after an initial evaluation or whose symptoms are unresponsive to a trial of glucocorticoids. A bronchoscopy can be used to assess for infection and, if necessary, obtain lung tissue to clarify the diagnosis [5,60,61]. However, the pathologic features for ICI pneumonitis are diverse and there is no one pathognomic finding. In one observational study that included 27 patients with ICI pneumonitis who underwent lung biopsy, histopathologic findings included cellular interstitial pneumonitis; organizing pneumonia; diffuse alveolar damage; or no abnormalities [52]. Interstitial inflammatory infiltrates included poorly formed granulomas or eosinophils.

Management — The management of pneumonitis is summarized in the American Society of Clinical Oncology (ASCO) guidelines table (table 5) [5]. There are no prospective clinical trials to define optimal management. Our treatment approach includes the following:

For asymptomatic, grade 1 pneumonitis (table 4), we generally withhold drug for two to four weeks with close follow-up. If symptoms arise or there is radiographic progression, glucocorticoids are appropriate.

Patients with grade 2 or higher pneumonitis should have their drug withheld and be treated using glucocorticoids with close follow-up. Additional immunosuppression may be used in patients with worsening of pneumonitis, with some evidence of benefit [63].

Outcomes from one observational study of 43 patients diagnosed with ICI pneumonitis provide some insight into the optimal management of this complication [52]. Overall, 15 of 17 asymptomatic patients (grade 1) were successfully managed by withholding the ICI, while 2 of the 17 and all 14 with grade 2 pneumonitis were successfully treated with glucocorticoids. All 12 patients with grade 3 or higher pneumonitis were initially treated with glucocorticoids. Five patients in this group required additional immunosuppression (infliximab with or without cyclophosphamide), but all five ultimately died. Of these five cases, infectious complications or progression of tumor appeared to be the proximal cause of death in four instances. The median starting dose of prednisone was 50 mg, and the median duration of treatment required was 68 days (range 20 to 154 days).

ENDOCRINOPATHIES — Inflammation of the pituitary, thyroid, or adrenal glands as a result of checkpoint blockade often presents with nonspecific symptoms such as nausea, headache, fatigue, and vision changes. The incidence of endocrinopathies has been difficult to precisely state due to variable methods of assessment, diagnosis, and monitoring in different clinical trials. The most common endocrinopathies are hypothyroidism, hyperthyroidism, and hypophysitis [64].

In a systematic review and meta-analysis that included 7551 patients in 38 randomized trials, the overall incidence of clinically significant endocrinopathies is approximately 10 percent of patients treated with ICIs [65]. The frequency of specific endocrinopathies and the relationship to different agents are discussed in this section.

The management of endocrine adverse events involving the thyroid, adrenal, pituitary, or endocrine pancreas is summarized in the American Society of Clinical Oncology (ASCO) guidelines table (table 6) [5].

Autoimmune thyroid disease — Thyroid function should be monitored prior to each dose of an ICI. Autoimmune thyroid disease can be manifested as primary hypothyroidism secondary to a destructive thyroiditis or as hyperthyroidism associated with Graves disease.

Hypothyroidism — Most commonly, thyroid disorders present with nonspecific symptoms such as fatigue. Since these symptoms can be vague, distinguishing primary thyroid disorders from secondary hypothyroidism (typically a result of hypophysitis) is critical to a thorough differential diagnosis.

Typically, a high thyroid-stimulating hormone (TSH) with low free thyroxine (T4) indicates primary hypothyroidism.

A low TSH with low free T4 indicates secondary hypothyroidism and likely hypophysitis. In such patients, levothyroxine should not be administered until patients have been evaluated for secondary adrenal insufficiency and, if diagnosed, treated with cortisol replacement. (See 'Hypophysitis' below and "Treatment of hypopituitarism", section on 'TSH deficiency'.)

Often a period of thyroiditis with transient hyperthyroidism (low TSH and high free T4) precedes longstanding hypothyroidism (high TSH and low free T4) [66]. For such patients, we do not suggest initial treatment of the hyperthyroid phase with anti-thyroid medication, since the phase is usually brief and almost invariably leads hypothyroidism. Instead, for select patients with significant symptoms attributed to hyperthyroidism, we suggest evaluating patients for the use of beta-blockers or other supportive medications. (See 'Hyperthyroidism' below.)

For patients treated with ipilimumab, nivolumab or pembrolizumab, atezolizumab, and the combination of nivolumab plus ipilimumab, the incidence rates for hypothyroidism were 3.8, 7, 3.9, and 13.2 percent, respectively [65]. There was no significant difference in incidence with nivolumab and pembrolizumab (6.5 and 7.9 percent, respectively).

Management of primary hypothyroidism typically involves replacement with thyroid hormone (levothyroxine) and endocrinology consultation.

Hyperthyroidism — Persistent primary hyperthyroidism is significantly less frequent than hypothyroidism and should be treated similarly to primary hyperthyroidism. The key to managing hyperthyroidism is to ensure it is persistent (eg, for at least two to three months) and not the initial transient hyperthyroid phase of thyroiditis that often leads to hypothyroidism. (See 'Hypothyroidism' above and "Graves' hyperthyroidism in nonpregnant adults: Overview of treatment".)

For patients treated with ipilimumab, nivolumab or pembrolizumab, atezolizumab, and the combination of nivolumab plus ipilimumab, the incidence rates for hyperthyroidism were 1.7, 3.2, 0.6, and 8 percent, respectively [65].

Hypophysitis

Clinical presentation and diagnosis — Typically, hypophysitis is manifested by clinical symptoms of fatigue and headache. The diagnosis is established by low levels of the hormones produced by the pituitary (adrenocorticotropic hormone [ACTH], TSH, follicle-stimulating hormone [FSH], luteinizing hormone [LH], growth hormone [GH], prolactin). (See "Clinical manifestations of hypopituitarism".)

Laboratory findings differentiate hypophysitis from primary adrenal insufficiency (manifested by low cortisol or inappropriate cortisol stimulation test and high ACTH) and primary hypothyroidism (manifested by low free T4 and high TSH). The diagnosis of hypophysitis is often supported radiographically by enhancement and swelling of the pituitary gland on magnetic resonance imaging of the brain (image 1) [67,68].

For patients treated with ipilimumab, nivolumab or pembrolizumab, atezolizumab, and the combination of nivolumab plus ipilimumab, the incidence rates for hypophysitis were 3.2, 0.4, <0.1, and 6.4 percent, respectively [65].

Management — In most patients, long-term supplementation of the affected hormones is necessary due to secondary hypothyroidism (treated with levothyroxine) or secondary adrenal insufficiency (treated with replacement doses of hydrocortisone, typically 20 mg each morning and 10 mg each evening). In some cases, patients can be successfully weaned from replacement steroids over time [69].

High doses of steroids are not typically needed to manage hypophysitis and have been associated with worse clinical outcomes [70]. However, for the rare patient who presents with pituitary compressive symptoms (eg, headache, double vision), a short course of high-dose corticosteroids (eg, prednisone 0.5 to 1 mg/kg daily for approximately one week) can be administered to alleviate such symptoms. (See "Treatment of hypopituitarism".)

Adrenal insufficiency — Adrenal insufficiency is a potentially life-threatening endocrinopathy which can cause dehydration, hypotension, and electrolyte imbalances (hyperkalemia, hyponatremia) and constitutes an emergency. Adrenal insufficiency is rare and has been reported in 0.7 percent of patients treated in randomized clinical trials [65].

When an adrenal crisis is suspected, intravenous glucocorticoids and immediate hospitalization is warranted. Consultation with an endocrinologist, aggressive hydration, and evaluation for sepsis are also critical. (See "Clinical manifestations of adrenal insufficiency in adults" and "Treatment of adrenal insufficiency in adults".)

Type 1 diabetes mellitus — Treatment with ICIs has been associated with acute onset of type 1 diabetes mellitus in approximately 0.2 to 0.9 percent of cases [65,71]. As examples, in several case series, patients typically presented with severe hyperglycemia or diabetic ketoacidosis; all required insulin therapy at diagnosis and remained insulin-dependent for diabetic control [71,72].

It is important to monitor glucose with each dose of immunotherapy. Patients who develop immunotherapy-induced type 1 diabetes mellitus are typically treated with insulin therapy. In contrast to other immune-related adverse events, treatment with glucocorticoids or other immunosuppressive agents is not effective in these patients, due to the almost complete destruction of the pancreatic beta cells by immunotherapy [71]. Immunotherapy-related diabetes may be suggested when the C-peptide level is low in the setting of hyperglycemia. (See "Diabetic ketoacidosis and hyperosmolar hyperglycemic state in adults: Treatment" and "Management of blood glucose in adults with type 1 diabetes mellitus".)

OPPORTUNISTIC INFECTIONS

Risk with immunosuppressive therapy — For most patients with no underlying pulmonary conditions receiving glucocorticoids for less than six weeks to treat an uncomplicated immune-related adverse event (irAE), we do not suggest Pneumocystis pneumonia (PCP) prophylaxis. Although PCP prophylaxis is often administered to patients on prolonged courses of glucocorticoids for other indications, it may not be necessary for those receiving them for an uncomplicated irAE. The incidence of opportunistic infections in patients with an irAE is low in observational studies, ranging between 2 and 7 percent [73,74]. Additionally, glucocorticoids in this patient population theoretically do not induce greater immunosuppression, but rather reduce immune activation from the ICI. This approach differs from the typical use of PCP prophylaxis in other HIV-uninfected patients. (See "Treatment and prevention of Pneumocystis pneumonia in patients without HIV", section on 'Prophylaxis'.)

However, prolonged immune suppression may put certain patients at risk for unusual or opportunistic infections. We offer PCP prophylaxis to patients receiving glucocorticoids for an irAE in the setting of combined chemotherapy plus immunotherapy; those with underlying pulmonary conditions receiving glucocorticoids for an uncomplicated irAE; or those with a complicated irAE (eg, those requiring longer than six weeks of glucocorticoids, or those not responsive to glucocorticoids requiring additional immunosuppressive therapy). The role of prophylactic antifungal or antiviral therapies in these patients requires further study.

In one series of 740 patients treated for advanced melanoma [73], approximately 80 percent of patients had been treated with ipilimumab, either alone or in combination with nivolumab. Serious infections were reported in 54 cases (7.3 percent). Specific infectious etiologies were bacterial, viral, fungal, and parasitic in 46, 6, 5, and 1 cases, respectively.

These infections were predominantly seen in association with glucocorticoids or infliximab. The incidence of serious infections was lower in those treated with nivolumab and pembrolizumab alone compared with ipilimumab or nivolumab plus ipilimumab, but this may have reflected the lower incidence of serious side effects necessitating immunosuppression.

Further details on the use of PCP prophylaxis in HIV-uninfected patients are discussed separately. (See "Treatment and prevention of Pneumocystis pneumonia in patients without HIV".)

LESS COMMON IMMUNE-RELATED ADVERSE EVENTS — Treatment with ICIs has also been associated with less common side effects in other organs. In some instances, these side effects have been severe or fatal.

Kidney — Acute kidney injury (AKI) is a rare but potentially serious complication of ICIs [75]. For patients with severe ICI-associated acute kidney injury (ICI-associated AKI), holding ICIs and treatment with glucocorticoids are indicated. One retrospective study suggested that patients with ICI nephritis treated with rapid glucocorticoid taper (over three weeks) had equivalent kidney recovery compared with those treated with longer glucocorticoid tapers (over six weeks) [76].

For patients whose kidney function improves, rechallenging with immunotherapy is a reasonable strategy. In one observational study of 429 patients with ICI-associated AKI, among the subset of patients who were subsequently rechallenged with immunotherapy, approximately 84 percent did not develop recurrent ICI-associated AKI [77]. (See 'Retreatment after prior toxicity' below.)

The estimated incidence of ICI-associated AKI is approximately 1.5 to 5 percent [78-80]. The most common reported underlying pathology is acute tubulointerstitial nephritis (image 2), but immune complex glomerulonephritis and thrombotic microangiopathy have also been observed [77,81-83].

The management of kidney toxicities is summarized in the American Society of Clinical Oncology (ASCO) guidelines table (table 7) [5].

Exocrine pancreas — The clinical presentation, diagnosis, and management of exocrine pancreatic toxicities associated with ICIs are discussed separately. (See "Hepatic, pancreatic, and rare gastrointestinal complications of immune checkpoint inhibitor therapy", section on 'Exocrine pancreas'.)

Neurologic — A wide range of neurologic syndromes have been associated with checkpoint blockade involving ipilimumab and programmed cell death receptor 1 (PD-1) pathway inhibitors [84-86]. Case series suggest that neurotoxicity occurs in approximately 1 to 14 percent of patients, with the highest rates associated with the use of combined immunotherapy using nivolumab plus ipilimumab [87-89]. As an example, in a review of approximately 9000 patients, the incidence of neurologic immune-related adverse events (irAEs) was 4 percent with cytotoxic T lymphocyte-associated antigen 4 (CTLA-4) blockade, 6 percent with PD-1 inhibitors, and 12 percent with combined CTLA-4 and PD-1 inhibition [90].

The most common symptoms are headache and peripheral sensory neuropathy. Neurologic complications associated with more severe toxicity include Guillain-Barré syndrome [91,92] and myasthenia gravis [1,93]. In some cases, myasthenia gravis may co-occur with inflammatory myositis and myocarditis and is associated with high fatality rates [94]. (See 'Cardiovascular toxicity' below and 'Rheumatologic and musculoskeletal' below.)

Other reported severe neurologic complications include posterior reversible encephalopathy syndrome [95], aseptic meningitis [96,97], enteric neuropathy [98], transverse myelitis [99], pancerebellitis [100], autoimmune encephalitis [101], and cranial and peripheral neuropathies. (See "Autoimmune (including paraneoplastic) encephalitis: Clinical features and diagnosis" and "Paraneoplastic syndromes affecting spinal cord, peripheral nerve, and muscle", section on 'Checkpoint inhibitor-associated neuropathies'.)

Neurologic irAEs typically develop within three months of starting ICI therapy, although the time of onset may vary [102]. Serious neurologic irAEs should be treated with glucocorticoids, even when the non-immunotherapy-induced version of the same neurologic disorder is not typically managed with glucocorticoids (such as Guillain-Barré syndrome). Consultation with neurology is indicated to consider additional treatment, such as plasmapheresis and intravenous immunoglobulin. Early recognition and intervention are key to reducing severity and duration of toxicity. As an example, in one observational study of 35 patients treated for neurologic irAEs, symptoms resolved in 26 patients (75 percent), with a median time to resolution of approximately one month [88].

The management of patients with immunotherapy-related neurologic toxicity is summarized in the ASCO guidelines table (table 8) [5].

Neurologic toxicities associated with other molecularly targeted and biologic agents are discussed separately. (See "Neurologic complications of cancer treatment with molecularly targeted and biologic agents".)

Cardiovascular toxicity — Cardiotoxicity may develop in the absence of a history of significant cardiac risk factors. Cardiac irAEs include myocarditis, pericarditis, heart failure, arrythmias, and vasculitis [103,104]. Venous thromboembolism may also be seen, although its relationship to ICIs is less clear.

Myocarditis – The time to onset was variable, but fatal myocarditis has been reported after a single treatment with the combination of nivolumab plus ipilimumab [105]. Observational studies also suggest that the incidence of myocarditis is higher in patients treated with the combination immunotherapy compared with single-agent immunotherapy [106].

High-dose corticosteroids (ie, prednisone 1 to 2 mg/kg/day) have been used to treat cardiac complications, but symptoms may progress in some cases despite aggressive therapy. Immediate transfer to a coronary care unit or, if available, cardiac transplant unit should be considered for patients with elevated troponin or conduction abnormalities. Concurrent skeletal muscle involvement (myositis) may also complicate therapy, as diaphragm involvement may lead to respiratory failure.

Patients without an immediate response to high-dose corticosteroids should be treated promptly with cardiac transplant rejection doses of steroids (methylprednisolone 1 g every day) and the addition of either mycophenolate, infliximab, or antithymocyte globulin. Abatacept [107] or alemtuzumab may be used as additional immunosuppression in life-threatening cases.

The management of ICI-associated myocarditis and other ICI-related cardiovascular toxicities is summarized in the ASCO guidelines table (table 9) [5].

Acute pericarditis – The management of acute pericarditis associated with the use of ICIs is discussed separately. (See "Pericardial disease associated with cancer: Management", section on 'Immune checkpoint inhibitor-associated pericarditis'.)

Hematologic — Various hematologic irAEs have been described in patients treated with ICIs. These include immune thrombocytopenia (ITP), autoimmune hemolytic anemia, hemophagocytic lymphohistiocytosis, red cell aplasia, neutropenia, acquired hemophilia A, and cryoglobulinemia, among others [108-120]. As with other irAEs, the standard approach is initial glucocorticoid treatment with the addition of other immunosuppressive agents if symptoms are glucocorticoid refractory. The management of common hematologic toxicities is summarized in the ASCO guidelines table (table 10) [5].

Select hematologic irAEs are discussed below:

Immune thrombocytopenia (ITP) – ITP is likely the most common hematologic irAE, although it occurs in approximately 1 percent of patients treated with ICIs. Peripheral blood smear and routine work-up for thrombocytopenia should be performed to rule out other rare causes of thrombocytopenia, including thrombotic thrombocytopenic purpura. Spontaneous resolution is common in mild cases. While glucocorticoids are effective for more severe cases of ITP, the addition of other immunosuppressive agents may also be necessary (such as intravenous immunoglobulin, thrombopoietin receptor agonists, and rituximab) [114,115]. Further details on the management of ITP are discussed separately. (See "Initial treatment of immune thrombocytopenia (ITP) in adults" and "Second-line and subsequent therapies for immune thrombocytopenia (ITP) in adults".)

Autoimmune hemolytic anemia – Autoimmune hemolytic anemia (AIHA) has been associated with the use of multiple immunotherapy agents (eg, atezolizumab, nivolumab, pembrolizumab, and ipilimumab) [116,117]. The treatment approach to immunotherapy-induced AIHA is similar to that of AIHA due to other drugs, which includes holding immunotherapy and initiation of glucocorticoid-based therapy. One observational study demonstrated that patients with immunotherapy-induced AIHA may have a negative direct antibody test (DAT), and most patients also resume immunotherapy without recurrent AIHA [118]. Further details regarding the diagnosis and management of this general condition are discussed separately. (See "Drug-induced hemolytic anemia".)

Hemophagocytic lymphohistiocytosis – Hemophagocytic lymphohistiocytosis (HLH) has been reported in patients receiving immunotherapy with nivolumab, ipilimumab, and/or pembrolizumab [119,120]. This is a rare but potentially fatal syndrome of excessive immune activation resulting in multi-organ failure, including cytopenias and bleeding. Further data are needed to confirm the association of either the PD-1 pathway or CTLA-4 inhibition with the development of HLH. Details regarding the treatment of HLH are discussed separately. (See "Treatment and prognosis of hemophagocytic lymphohistiocytosis".)

Eye — CTLA-4 blockade with ipilimumab has been associated with eye inflammation, which can be manifested by episcleritis, conjunctivitis, uveitis, or orbital inflammation. The incidence is less than 1 percent, and symptoms can include photophobia, pain, dryness of the eyes, and blurred vision.

Intraocular inflammation (uveitis) following treatment with pembrolizumab or nivolumab is a rare but clinically important event described in approximately 1 percent of treated patients. Although the available data are limited, the risk of eye disorders may be aggravated when drugs of both ICI classes are combined. (See "Ocular side effects of systemically administered chemotherapy", section on 'Anti-PD-1 and PD-L1 agents'.)

An ophthalmology consultation is recommended, and treatment with topical glucocorticoids (eg, 1 percent prednisolone acetate suspension) may be helpful. Oral glucocorticoids can be used for severe (grade 3/4) or refractory cases. The management of eye toxicity is summarized in the ASCO guidelines table and discussed in detail separately (table 11). (See "Ocular side effects of systemically administered chemotherapy", section on 'Ipilimumab'.)

Rheumatologic and musculoskeletal — A wide range of rheumatologic toxicities has been observed with ICIs. These include myositis, inflammatory arthritis, salivary gland dysfunction (sicca syndrome), and vasculitis, among others [121-125]. The incidence of these side effects has not been clearly determined. (See "Rheumatologic complications of checkpoint inhibitor immunotherapy".)

Myositis can also be seen with ICIs and is occasionally severe/fatal. Given the possibility of severe cases, Health Canada issued a safety alert urging recognition and management of myositis [126]. Patients who have significant myositis should be evaluated for myocarditis since these syndromes commonly manifest together. (See 'Cardiovascular toxicity' above.)

The management of musculoskeletal toxicities is summarized in the ASCO guidelines table (table 12) [5].

RETREATMENT AFTER PRIOR TOXICITY — Retreatment with ICIs may be safely offered to most patients with significant immune-related adverse events (irAEs) during initial treatment that consists of either a cytotoxic T lymphocyte-associated antigen 4 (CTLA-4) inhibitor and/or a programmed cell death receptor 1 (PD-1)/programmed cell death ligand 1 (PD-L1) inhibitor [127-134].

The choice to retreat is dependent on multiple factors, including the severity and nature of the initial irAE, its degree of responsiveness to systemic immunosuppression, and the availability of alternative treatment options. Clinicians should also provide a careful risk-benefit discussion to patients who are candidates for retreatment.

Data are limited on the specific patient populations who should not be offered retreatment with ICIs, and clinical judgment is necessary. As examples:

Patients who survive a frequently fatal irAE (eg, myocarditis) are not routinely offered retreatment.

Similarly, retreatment is discouraged in those actively receiving glucocorticoid doses equivalent to prednisone 10 mg daily or higher for treatment of an initial irAE, as the concurrent use of glucocorticoids is associated with reduced efficacy of immunotherapy [135]. (See "Initial management of advanced non-small cell lung cancer lacking a driver mutation", section on 'Impact of steroids on efficacy of immunotherapy'.)

Data are also limited for the oncologic benefits of retreatment in those experiencing an initial irAE, particularly in those who have had a complete or sustained response to the initial regimen and do not require further intervention. (See "Systemic treatment of metastatic melanoma lacking a BRAF mutation", section on 'What is the optimal duration of immunotherapy?'.)

For patients who are candidates for retreatment with ICIs, dose reductions are not recommended with retreatment, as this approach has not been assessed in clinical trials. The optimal choice of agent also varies based on clinical practice. Patients who experience severe toxicity from an initial regimen containing a CTLA-4 inhibitor are generally offered retreatment with single-agent PD-1 or PD-L1 inhibitors rather than rechallenging with a CTLA-4 inhibitor.

In studies of patients who are retreated with various classes of ICIs (table 1), the rate of recurrent irAEs for PD-1 or PD-L1 inhibitors ranges between 18 and 55 percent; however, a majority of these irAEs are mild and managed successfully with immunosuppressive therapy [127,129-132,136]. In patients with irAEs from combination PD-1 plus CTLA-4 inhibition, approximately 20 percent had recurrence of irAE with resumption of single agent PD-1 inhibitors [130]. In contrast, retreatment with CTLA-4 inhibitors are associated with the highest rates of recurrent irAEs (up to 88 percent in one study [127]).

The approach to resuming immunotherapy after ICI colitis and ICI hepatitis are discussed separately. (See "Immune checkpoint inhibitor colitis", section on 'Resuming immune checkpoint inhibitors' and "Hepatic, pancreatic, and rare gastrointestinal complications of immune checkpoint inhibitor therapy", section on 'Resuming immunotherapy after prior toxicity'.)

PATIENTS WITH VULNERABILITIES TO IMMUNOTHERAPY TOXICITIES

Pre-existing autoimmune disease — In patients with pre-existing autoimmune disease, there are limited data on the safety and efficacy of ICIs [32,137]. Such patients were excluded from clinical trials evaluating immunotherapy due to concerns about exacerbating underlying autoimmune disease or immune-related adverse events (irAEs) [138]. Observational studies suggest that most patients with autoimmune disease can safely receive ICIs [32,129,139-144]. However, compared with those without autoimmune disease, these patients may be at higher risk for specific irAEs (eg, immune-mediated colitis in those with inflammatory bowel disease [IBD], cardiovascular irAEs [145]) or discontinuing immunotherapy due to irAEs [32]; they may also be at high risk for developing an exacerbation of their underlying autoimmune disorders [129,138-140,142,146].

Clinicians should offer such patients a cautious risk-benefit discussion prior to initiating immunotherapy and evaluate various factors including the effectiveness of immunotherapy for the underlying malignancy; performance status and comorbidities; and type and severity of the autoimmune condition. Patients should also be evaluated for contraindications to immunotherapy (eg, poorly controlled autoimmune disease, active immunosuppression using the equivalent of prednisone 10 mg daily or higher) that require alternative therapies for their cancer [147]. Immunotherapy should be used with extreme caution or avoided in patients with potentially life-threatening autoimmune conditions [148]. We also recommend discussion of treatment options with both the patient and the clinician responsible for treating their autoimmune condition before determining a specific plan of action. Further details on the general approach to managing irAEs in patients with specific autoimmune disorders are discussed separately. (See "Rheumatologic complications of checkpoint inhibitor immunotherapy", section on 'General principles of evaluation and management' and "Systemic treatment of metastatic melanoma with BRAF and other molecular alterations", section on 'Defining immunotherapy eligibility'.)

In a prospective observational cohort study of 4367 patients with advanced melanoma, 415 patients (10 percent) with autoimmune disease were identified [32]. This cohort included 227 patients with rheumatologic disease (rheumatoid arthritis, systemic lupus erythematosus, and others); 143 patients with endocrine disease (hypo- or hyperthyroidism, Graves disease); 155 patients with IBD; 8 patients with "other" autoimmune conditions, and 20 with multiple autoimmune conditions. Of note, patients with autoimmune disease and active immunosuppression were more likely to receive alternative initial therapy (eg, BRAF plus MEK inhibitors) rather than immunotherapy.

Among the 55 percent who received immunotherapy (228 patients), the incidence of grade ≥3 irAEs was similar to that seen in a cohort of patients with advanced melanoma and no autoimmune disease, regardless of the choice of immunotherapy (30 percent each for a cytotoxic T lymphocyte-associated antigen 4 [CTLA-4] inhibitor; 17 versus 13 percent for a programmed cell death receptor 1 [PD-1] inhibitor; and 44 versus 48 percent for combination immunotherapy). Among those treated with PD-1 inhibitors, patients with autoimmune disease were more likely to discontinue immunotherapy due to irAEs compared with those without autoimmune disease (17 versus 9 percent). Patients with IBD treated with PD-1 inhibitors were more likely to develop immunotherapy-induced colitis (6 of 31 patients [19 percent]) versus those with other (non-IBD) autoimmune diseases (3 percent) and those without autoimmune disease (2 percent), which is consistent with prior studies [138]. However, data on acute flares of patients' pre-existing autoimmune condition were not reported.

Objective response rates and overall survival were similar for those with and without autoimmune disease, although this could be biased by the imbalance of baseline treatment characteristics between the two groups. The impact of other immunosuppressive agents (eg, infliximab or vedolizumab) on clinical outcomes is not known. In addition, approximately one-third of the study population was comprised of patients with thyroid disease (for whom there is less concern regarding irAEs). However, this study did not include patients with more rare, potentially fatal autoimmune conditions/irAEs (eg, myositis, myasthenia gravis, and Guillain-Barré syndrome) [3]. (See 'Relationship between immunotherapy toxicities and efficacy' above.)

Older adult patients — Data suggest that ICIs have similar efficacy and toxicity in older adults as in younger adults, and that chronologic age alone should not preclude the use of these agents [149-153].

A meta-analysis of nine randomized trials that compared nivolumab, pembrolizumab, or atezolizumab with chemotherapy or targeted therapy in solid tumors (non-small cell lung cancer, melanoma, renal cell carcinoma, head and neck cancer) analyzed efficacy in 5458 patients [149]. There was a consistent overall survival advantage for immunotherapy across all trials (hazard ratio [HR] 0.69, 95% CI 0.63-0.74), and there was no difference between those <65 years (HR 0.68, 95% CI 0.61-0.75) and those ≥65 years (HR 0.64, 95% CI 0.54-0.76). An analysis of progression-free survival based on age was available in four trials. There was no significant difference between the two age cohorts (HR 0.73, 95% CI 0.61-0.88, and HR 0.74, 95% CI 0.6-0.92, respectively). However, limitations of this meta-analysis include the potential lack of applicability to those with an impaired performance status or significant comorbidity, the limited number of patients 75 years or older, and the lack of relative toxicity data in the older adult population versus younger patients.

Another retrospective study in approximately 900 patients age 80 years or older treated with ICIs showed similar efficacy and severe toxicity rates compared with younger patients [152]. Of note, the oldest population (≥90 years old) frequently discontinued therapy due to irAEs, even though these were often low grade.

SOCIETY GUIDELINE LINKS — Links to society and government-sponsored guidelines from selected countries and regions around the world are provided separately. (See "Society guideline links: Management of toxicities due to checkpoint inhibitor immunotherapy".)

SUMMARY AND RECOMMENDATIONS

Definitions – Immune checkpoint inhibitors (ICIs), also known as checkpoint inhibitor immunotherapy, are antibodies that target cytotoxic T lymphocyte-associated antigen 4 (CTLA-4), programmed cell death receptor 1 (PD-1), and programmed cell death ligand 1 (PD-L1). (See 'Introduction' above.)

ICIs are widely used in the treatment of various types of cancer.

Despite important clinical benefits, ICIs are associated with a unique spectrum of side effects known as immune-related adverse events (irAEs).

Impact of immunotherapy agent on irAEs – IrAEs are more common with agents that target CTLA-4 (ipilimumab, tremelimumab) than agents that target PD-1 (nivolumab, pembrolizumab, cemiplimab, and dostarlimab) or PD-L1 (atezolizumab, durvalumab, avelumab (table 1)). The combination of nivolumab plus ipilimumab is associated with more toxicity than either agent alone. (See 'General principles' above.)

Common irAEs – Although many sites can be affected, the most common and important irAEs are:

Dermatologic (see "Cutaneous immune-related adverse events associated with immune checkpoint inhibitors")

Diarrhea/colitis (see "Immune checkpoint inhibitor colitis")

Hepatotoxicity (see "Hepatic, pancreatic, and rare gastrointestinal complications of immune checkpoint inhibitor therapy", section on 'Hepatotoxicity')

Pneumonitis (see 'Pneumonitis' above)

Endocrinopathies (see 'Endocrinopathies' above)

Less commonly affected sites – Less commonly affected sites include the kidney, exocrine pancreas, central nervous system, cardiovascular, hematologic, eye, and rheumatologic and musculoskeletal systems. (See 'Less common immune-related adverse events' above.)

Treatment approach – Rapid identification of irAEs and prompt initiation of immunosuppression can optimize outcomes. In general, treatment of moderate or severe irAEs requires interruption of ICIs and immunosuppression with glucocorticoids. Treatment is based on site of the irAE and the severity of the observed toxicity (see 'General principles' above):

Grade 1 irAEs – For most patients with grade 1 irAEs (asymptomatic or mild symptoms), immunotherapy may be continued, and patients may be closely monitored for worsening irAEs and/or managed symptomatically.

Grade 2 irAEs – For most patients with grade 2 (moderate) immune-mediated toxicities, treatment with immunotherapy should be withheld and should not be resumed until symptoms or toxicity is grade 1 or less. Glucocorticoids (prednisone 0.5 mg/kg/day or equivalent) should be started if symptoms do not resolve within one week.

-Grade 2 endocrinopathies – For those with grade 2 immune-mediated endocrinopathies, immunotherapy may be withheld until hormone replacement is initiated and subsequently resumed once acute symptoms have resolved and (in the event of adrenal insufficiency or hypophysitis) patients are receiving adequate adrenal glucocorticoid replacement.

Grade 3 or 4 irAEs – For patients experiencing grade 3 or 4 (severe or life-threatening) immune-mediated toxicities, treatment with the immunotherapy is usually permanently discontinued. High doses of glucocorticoids (prednisone 1 to 2 mg/kg/day or equivalent) should be given. When symptoms subside to grade 1 or less, glucocorticoids can be gradually tapered over at least one month.

Refractory toxicity – If glucocorticoids are not effective, our approach is to administer an additional immunosuppressive agent (eg, infliximab, vedolizumab, mycophenolate mofetil, among other options).

Communication – Frequent and consistent communication between patients, caregivers, and the clinical team is vital to successful irAE management.

Immunotherapy plus other agents – Immunotherapy in combination with other systemic agents may result in varied toxicity profiles. Clinicians should assess and treat the toxicities associated with each individual agent and remain aware of the potential for development of novel toxicities. (See 'Combining or sequencing immunotherapy with other therapies' above.)

Indications for PCP prophylaxis – For patients with no underlying pulmonary conditions receiving glucocorticoids for less than six weeks to treat an uncomplicated irAE, we suggest observation rather than pharmacologic Pneumocystis pneumonia (PCP) prophylaxis (Grade 2C).

We offer PCP prophylaxis to patients receiving glucocorticoids for an irAE in the setting of combined chemotherapy plus immunotherapy; those with underlying pulmonary conditions receiving glucocorticoids for an uncomplicated irAE; or those with a complicated irAE (eg, those requiring longer than six weeks of glucocorticoids or additional immunosuppressive therapy). (See 'Opportunistic infections' above.)

Pre-existing autoimmune disease

In patients with pre-existing autoimmune disease receiving immunotherapy, limited observational data suggest a similar incidence of overall irAEs compared with those without autoimmune disease, but a potentially higher risk for developing specific irAEs (eg, immune-mediated colitis in patients with inflammatory bowel disease) and/or discontinuing therapy. (See 'Pre-existing autoimmune disease' above.)

Although data suggest that most patients with autoimmune disease can safely receive immunotherapy, clinicians should offer a cautious risk-benefit discussion prior to initiating therapy, evaluate for contraindications (eg, poorly controlled autoimmune disease, active immunosuppression) that require alternative therapies, and involve the clinician treating the patient's autoimmune condition.

Retreatment after prior toxicity – Retreatment with ICIs may be safely offered to most patients with significant irAEs during initial treatment with immunotherapy, after a risk-benefit discussion. The choice to retreat is dependent on multiple factors, including the severity and nature of the initial irAE, its degree of responsiveness to systemic immunosuppression, the clinical response to the initial immunotherapy regimen, and the availability of alternative treatment options. (See 'Retreatment after prior toxicity' above.)

ACKNOWLEDGMENT — The UpToDate editorial staff acknowledges Jedd Wolchok, MD, PhD, who contributed to earlier versions of this topic review.

  1. Naidoo J, Page DB, Li BT, et al. Toxicities of the anti-PD-1 and anti-PD-L1 immune checkpoint antibodies. Ann Oncol 2015; 26:2375.
  2. Champiat S, Lambotte O, Barreau E, et al. Management of immune checkpoint blockade dysimmune toxicities: a collaborative position paper. Ann Oncol 2016; 27:559.
  3. Wang DY, Salem JE, Cohen JV, et al. Fatal Toxic Effects Associated With Immune Checkpoint Inhibitors: A Systematic Review and Meta-analysis. JAMA Oncol 2018; 4:1721.
  4. Fujiwara Y, Horita N, Adib E, et al. Treatment-related adverse events, including fatal toxicities, in patients with solid tumours receiving neoadjuvant and adjuvant immune checkpoint blockade: a systematic review and meta-analysis of randomised controlled trials. Lancet Oncol 2024; 25:62.
  5. Schneider BJ, Naidoo J, Santomasso BD, et al. Management of Immune-Related Adverse Events in Patients Treated With Immune Checkpoint Inhibitor Therapy: ASCO Guideline Update. J Clin Oncol 2021; 39:4073.
  6. https://www.sitcancer.org/research/cancer-immunotherapy-guidelines. Society for Immunotherapy of Cancer (SITC) Cancer Immunotherapy Guidelines (Accessed on January 10, 2022).
  7. Weber JS, Hodi FS, Wolchok JD, et al. Safety Profile of Nivolumab Monotherapy: A Pooled Analysis of Patients With Advanced Melanoma. J Clin Oncol 2017; 35:785.
  8. Horvat TZ, Adel NG, Dang TO, et al. Immune-Related Adverse Events, Need for Systemic Immunosuppression, and Effects on Survival and Time to Treatment Failure in Patients With Melanoma Treated With Ipilimumab at Memorial Sloan Kettering Cancer Center. J Clin Oncol 2015; 33:3193.
  9. Lefort F, Dalban C, Gross-Goupil M, et al. Association of Corticosteroids With Survival Outcomes in Patients With Metastatic Renal Cell Carcinoma Treated With Nivolumab. JAMA Oncol 2023; 9:1295.
  10. Bai X, Hu J, Betof Warner A, et al. Early Use of High-Dose Glucocorticoid for the Management of irAE Is Associated with Poorer Survival in Patients with Advanced Melanoma Treated with Anti-PD-1 Monotherapy. Clin Cancer Res 2021; 27:5993.
  11. van Not OJ, Verheijden RJ, van den Eertwegh AJM, et al. Association of Immune-Related Adverse Event Management With Survival in Patients With Advanced Melanoma. JAMA Oncol 2022; 8:1794.
  12. Maher VE, Fernandes LL, Weinstock C, et al. Analysis of the Association Between Adverse Events and Outcome in Patients Receiving a Programmed Death Protein 1 or Programmed Death Ligand 1 Antibody. J Clin Oncol 2019; 37:2730.
  13. Eggermont AMM, Kicinski M, Blank CU, et al. Association Between Immune-Related Adverse Events and Recurrence-Free Survival Among Patients With Stage III Melanoma Randomized to Receive Pembrolizumab or Placebo: A Secondary Analysis of a Randomized Clinical Trial. JAMA Oncol 2020; 6:519.
  14. Topalian SL, Hodi FS, Brahmer JR, et al. Five-Year Survival and Correlates Among Patients With Advanced Melanoma, Renal Cell Carcinoma, or Non-Small Cell Lung Cancer Treated With Nivolumab. JAMA Oncol 2019; 5:1411.
  15. Akamatsu H, Murakami E, Oyanagi J, et al. Immune-Related Adverse Events by Immune Checkpoint Inhibitors Significantly Predict Durable Efficacy Even in Responders with Advanced Non-Small Cell Lung Cancer. Oncologist 2020; 25:e679.
  16. Suo A, Chan Y, Beaulieu C, et al. Anti-PD1-Induced Immune-Related Adverse Events and Survival Outcomes in Advanced Melanoma. Oncologist 2020; 25:438.
  17. Kichenadasse G, Miners JO, Mangoni AA, et al. Multiorgan Immune-Related Adverse Events During Treatment With Atezolizumab. J Natl Compr Canc Netw 2020; 18:1191.
  18. Bottlaender L, Amini-Adle M, Maucort-Boulch D, et al. Cutaneous adverse events: a predictor of tumour response under anti-PD-1 therapy for metastatic melanoma, a cohort analysis of 189 patients. J Eur Acad Dermatol Venereol 2020; 34:2096.
  19. Schweizer C, Schubert P, Rutzner S, et al. Prospective evaluation of the prognostic value of immune-related adverse events in patients with non-melanoma solid tumour treated with PD-1/PD-L1 inhibitors alone and in combination with radiotherapy. Eur J Cancer 2020; 140:55.
  20. Shankar B, Zhang J, Naqash AR, et al. Multisystem Immune-Related Adverse Events Associated With Immune Checkpoint Inhibitors for Treatment of Non-Small Cell Lung Cancer. JAMA Oncol 2020; 6:1952.
  21. Zou F, Abu-Sbeih H, Ma W, et al. Association of Chronic Immune-Mediated Diarrhea and Colitis With Favorable Cancer Response. J Natl Compr Canc Netw 2020; 19:700.
  22. Atkins MB, Mier JW, Parkinson DR, et al. Hypothyroidism after treatment with interleukin-2 and lymphokine-activated killer cells. N Engl J Med 1988; 318:1557.
  23. D'Angelo SP, Larkin J, Sosman JA, et al. Efficacy and Safety of Nivolumab Alone or in Combination With Ipilimumab in Patients With Mucosal Melanoma: A Pooled Analysis. J Clin Oncol 2017; 35:226.
  24. Socinski MA, Jotte RM, Cappuzzo F, et al. Pooled analyses of immune-related adverse events (irAEs) and efficacy from the phase 3 trials IMpower130, IMpower132, and IMpower150. J Oncol 2021; 39;15S.
  25. Martini DJ, Goyal S, Liu Y, et al. Immune-Related Adverse Events as Clinical Biomarkers in Patients with Metastatic Renal Cell Carcinoma Treated with Immune Checkpoint Inhibitors. Oncologist 2021; 26:e1742.
  26. Foster CC, Couey MA, Kochanny SE, et al. Immune-related adverse events are associated with improved response, progression-free survival, and overall survival for patients with head and neck cancer receiving immune checkpoint inhibitors. Cancer 2021; 127:4565.
  27. Reck M, Ciuleanu TE, Cobo M, et al. First-line nivolumab plus ipilimumab with two cycles of chemotherapy versus chemotherapy alone (four cycles) in advanced non-small-cell lung cancer: CheckMate 9LA 2-year update. ESMO Open 2021; 6:100273.
  28. Fukushima T, Morimoto M, Kobayashi S, et al. Association Between Immune-Related Adverse Events and Survival in Patients with Hepatocellular Carcinoma Treated With Atezolizumab Plus Bevacizumab. Oncologist 2023; 28:e526.
  29. Ascierto PA, Simeone E, Sileni VC, et al. Clinical experience with ipilimumab 3 mg/kg: real-world efficacy and safety data from an expanded access programme cohort. J Transl Med 2014; 12:116.
  30. Nigro O, Pinotti G, De Galitiis F, et al. Late immune-related adverse events in long-term responders to PD-1/PD-L1 checkpoint inhibitors: A multicentre study. Eur J Cancer 2020; 134:19.
  31. Robert C, Hwu WJ, Hamid O, et al. Long-term safety of pembrolizumab monotherapy and relationship with clinical outcome: A landmark analysis in patients with advanced melanoma. Eur J Cancer 2021; 144:182.
  32. van der Kooij MK, Suijkerbuijk KPM, Aarts MJB, et al. Safety and Efficacy of Checkpoint Inhibition in Patients With Melanoma and Preexisting Autoimmune Disease : A Cohort Study. Ann Intern Med 2021; 174:641.
  33. Unger JM, Vaidya R, Albain KS, et al. Sex Differences in Risk of Severe Adverse Events in Patients Receiving Immunotherapy, Targeted Therapy, or Chemotherapy in Cancer Clinical Trials. J Clin Oncol 2022; 40:1474.
  34. McCulloch JA, Davar D, Rodrigues RR, et al. Intestinal microbiota signatures of clinical response and immune-related adverse events in melanoma patients treated with anti-PD-1. Nat Med 2022; 28:545.
  35. Smithy JW, Faleck DM, Postow MA. Facts and Hopes in Prediction, Diagnosis, and Treatment of Immune-Related Adverse Events. Clin Cancer Res 2022; 28:1250.
  36. Schmidt EV, Chisamore MJ, Chaney MF, et al. Assessment of Clinical Activity of PD-1 Checkpoint Inhibitor Combination Therapies Reported in Clinical Trials. JAMA Netw Open 2020; 3:e1920833.
  37. Magee DE, Hird AE, Klaassen Z, et al. Adverse event profile for immunotherapy agents compared with chemotherapy in solid organ tumors: a systematic review and meta-analysis of randomized clinical trials. Ann Oncol 2020; 31:50.
  38. Ribas A, Hodi FS, Callahan M, et al. Hepatotoxicity with combination of vemurafenib and ipilimumab. N Engl J Med 2013; 368:1365.
  39. Minor DR, Puzanov I, Callahan MK, et al. Severe gastrointestinal toxicity with administration of trametinib in combination with dabrafenib and ipilimumab. Pigment Cell Melanoma Res 2015; 28:611.
  40. Ribas A, Butler M, Lutzky J, et al. Phase I study combining anti-PD-L1 (MEDI4736) with BRAF (dabrafenib) and/or MEK (trametinib) inhibitors in advanced melanoma. J Clin Oncol 2015; 33S: ASCO #3003.
  41. Sullivan RJ, Hamid O, Gonzalez R, et al. Atezolizumab plus cobimetinib and vemurafenib in BRAF-mutated melanoma patients. Nat Med 2019; 25:929.
  42. Ascierto PA, Ferrucci PF, Fisher R, et al. Dabrafenib, trametinib and pembrolizumab or placebo in BRAF-mutant melanoma. Nat Med 2019; 25:941.
  43. Ribas A, Lawrence D, Atkinson V, et al. Combined BRAF and MEK inhibition with PD-1 blockade immunotherapy in BRAF-mutant melanoma. Nat Med 2019; 25:936.
  44. Harding JJ, Pulitzer M, Chapman PB. Vemurafenib sensitivity skin reaction after ipilimumab. N Engl J Med 2012; 366:866.
  45. Anscher MS, Arora S, Weinstock C, et al. Association of Radiation Therapy With Risk of Adverse Events in Patients Receiving Immunotherapy: A Pooled Analysis of Trials in the US Food and Drug Administration Database. JAMA Oncol 2022; 8:232.
  46. Zhou X, Yao Z, Bai H, et al. Treatment-related adverse events of PD-1 and PD-L1 inhibitor-based combination therapies in clinical trials: a systematic review and meta-analysis. Lancet Oncol 2021; 22:1265.
  47. Avelumab injection. United States Prescribing Information. US National Library of Medicine. Available online at: http://www.accessdata.fda.gov/drugsatfda_docs/label/2017/761049s000lbl.pdf (Accessed on April 08, 2019).
  48. Summary Safety Review - Opdivo (nivolumab) - Assessing the Potential Risks of Certain Blood Disorders, and Cytokine Release and Tumor Lysis Syndromes https://hpr-rps.hres.ca/reg-content/summary-safety-review-detail.php?lang=en&linkID=SSR00267 (Accessed on July 19, 2021).
  49. Zhao L, Yang Y, Li W, et al. Nivolumab-induced cytokine-release syndrome in relapsed/refractory Hodgkin's lymphoma: a case report and literature review. Immunotherapy 2018; 10:913.
  50. Nishino M, Giobbie-Hurder A, Hatabu H, et al. Incidence of Programmed Cell Death 1 Inhibitor-Related Pneumonitis in Patients With Advanced Cancer: A Systematic Review and Meta-analysis. JAMA Oncol 2016; 2:1607.
  51. Nishino M, Ramaiya NH, Awad MM, et al. PD-1 Inhibitor-Related Pneumonitis in Advanced Cancer Patients: Radiographic Patterns and Clinical Course. Clin Cancer Res 2016; 22:6051.
  52. Naidoo J, Wang X, Woo KM, et al. Pneumonitis in Patients Treated With Anti-Programmed Death-1/Programmed Death Ligand 1 Therapy. J Clin Oncol 2017; 35:709.
  53. Delaunay M, Cadranel J, Lusque A, et al. Immune-checkpoint inhibitors associated with interstitial lung disease in cancer patients. Eur Respir J 2017; 50.
  54. Khunger M, Rakshit S, Pasupuleti V, et al. Incidence of Pneumonitis With Use of Programmed Death 1 and Programmed Death-Ligand 1 Inhibitors in Non-Small Cell Lung Cancer: A Systematic Review and Meta-Analysis of Trials. Chest 2017; 152:271.
  55. Chen X, Zhang Z, Hou X, et al. Immune-related pneumonitis associated with immune checkpoint inhibitors in lung cancer: a network meta-analysis. J Immunother Cancer 2020; 8.
  56. Tiu BC, Zubiri L, Iheke J, et al. Real-world incidence and impact of pneumonitis in patients with lung cancer treated with immune checkpoint inhibitors: a multi-institutional cohort study. J Immunother Cancer 2022; 10.
  57. Atchley WT, Alvarez C, Saxena-Beem S, et al. Immune Checkpoint Inhibitor-Related Pneumonitis in Lung Cancer: Real-World Incidence, Risk Factors, and Management Practices Across Six Health Care Centers in North Carolina. Chest 2021; 160:731.
  58. Owen CN, Bai X, Quah T, et al. Delayed immune-related adverse events with anti-PD-1-based immunotherapy in melanoma. Ann Oncol 2021; 32:917.
  59. Shibaki R, Akamatsu H, Fujimoto M, et al. Nivolumab induced radiation recall pneumonitis after two years of radiotherapy. Ann Oncol 2017; 28:1404.
  60. Friedman CF, Proverbs-Singh TA, Postow MA. Treatment of the Immune-Related Adverse Effects of Immune Checkpoint Inhibitors: A Review. JAMA Oncol 2016; 2:1346.
  61. Marin-Acevedo JA, Chirila RM, Dronca RS. Immune Checkpoint Inhibitor Toxicities. Mayo Clin Proc 2019; 94:1321.
  62. Grimm MO, Bex A, De Santis M, et al. Safe Use of Immune Checkpoint Inhibitors in the Multidisciplinary Management of Urological Cancer: The European Association of Urology Position in 2019. Eur Urol 2019; 76:368.
  63. Beattie J, Rizvi H, Fuentes P, et al. Success and failure of additional immune modulators in steroid-refractory/resistant pneumonitis related to immune checkpoint blockade. J Immunother Cancer 2021; 9.
  64. Abdel-Rahman O, ElHalawani H, Fouad M. Risk of endocrine complications in cancer patients treated with immune check point inhibitors: a meta-analysis. Future Oncol 2016; 12:413.
  65. Barroso-Sousa R, Barry WT, Garrido-Castro AC, et al. Incidence of Endocrine Dysfunction Following the Use of Different Immune Checkpoint Inhibitor Regimens: A Systematic Review and Meta-analysis. JAMA Oncol 2018; 4:173.
  66. Muir CA, Clifton-Bligh RJ, Long GV, et al. Thyroid Immune-related Adverse Events Following Immune Checkpoint Inhibitor Treatment. J Clin Endocrinol Metab 2021; 106:e3704.
  67. Blansfield JA, Beck KE, Tran K, et al. Cytotoxic T-lymphocyte-associated antigen-4 blockage can induce autoimmune hypophysitis in patients with metastatic melanoma and renal cancer. J Immunother 2005; 28:593.
  68. Dillard T, Yedinak CG, 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.
  69. Sarnaik AA, Yu B, Yu D, et al. Extended dose ipilimumab with a peptide vaccine: immune correlates associated with clinical benefit in patients with resected high-risk stage IIIc/IV melanoma. Clin Cancer Res 2011; 17:896.
  70. Faje AT, Lawrence D, Flaherty K, et al. High-dose glucocorticoids for the treatment of ipilimumab-induced hypophysitis is associated with reduced survival in patients with melanoma. Cancer 2018; 124:3706.
  71. Clotman K, Janssens K, Specenier P, et al. Programmed Cell Death-1 Inhibitor-Induced Type 1 Diabetes Mellitus. J Clin Endocrinol Metab 2018; 103:3144.
  72. Hughes J, Vudattu N, Sznol M, et al. Precipitation of autoimmune diabetes with anti-PD-1 immunotherapy. Diabetes Care 2015; 38:e55.
  73. Del Castillo M, Romero FA, Argüello E, et al. The Spectrum of Serious Infections Among Patients Receiving Immune Checkpoint Blockade for the Treatment of Melanoma. Clin Infect Dis 2016; 63:1490.
  74. Cook MR, Shah NJ, Blackburn M, et al. Opportunistic Infection Outcomes in Patients Receiving Prolonged Corticosteroids for Immune Related Adverse Events. J Immunother Cancer 2019; Abstract Supplement.
  75. Manohar S, Kompotiatis P, Thongprayoon C, et al. Programmed cell death protein 1 inhibitor treatment is associated with acute kidney injury and hypocalcemia: meta-analysis. Nephrol Dial Transplant 2019; 34:108.
  76. Lee MD, Seethapathy H, Strohbehn IA, et al. Rapid corticosteroid taper versus standard of care for immune checkpoint inhibitor induced nephritis: a single-center retrospective cohort study. J Immunother Cancer 2021; 9.
  77. Gupta S, Short SAP, Sise ME, et al. Acute kidney injury in patients treated with immune checkpoint inhibitors. J Immunother Cancer 2021; 9.
  78. Cortazar FB, Kibbelaar ZA, Glezerman IG, et al. Clinical Features and Outcomes of Immune Checkpoint Inhibitor-Associated AKI: A Multicenter Study. J Am Soc Nephrol 2020; 31:435.
  79. Kitchlu A, Jhaveri KD, Wadhwani S, et al. A Systematic Review of Immune Checkpoint Inhibitor-Associated Glomerular Disease. Kidney Int Rep 2021; 6:66.
  80. Espi M, Teuma C, Novel-Catin E, et al. Renal adverse effects of immune checkpoints inhibitors in clinical practice: ImmuNoTox study. Eur J Cancer 2021; 147:29.
  81. Cortazar FB, Marrone KA, Troxell ML, et al. Clinicopathological features of acute kidney injury associated with immune checkpoint inhibitors. Kidney Int 2016; 90:638.
  82. Izzedine H, Mateus C, Boutros C, et al. Renal effects of immune checkpoint inhibitors. Nephrol Dial Transplant 2017; 32:936.
  83. Mamlouk O, Selamet U, Machado S, et al. Nephrotoxicity of immune checkpoint inhibitors beyond tubulointerstitial nephritis: single-center experience. J Immunother Cancer 2019; 7:2.
  84. Wick W, Hertenstein A, Platten M. Neurological sequelae of cancer immunotherapies and targeted therapies. Lancet Oncol 2016; 17:e529.
  85. Kao JC, Liao B, Markovic SN, et al. Neurological Complications Associated With Anti-Programmed Death 1 (PD-1) Antibodies. JAMA Neurol 2017; 74:1216.
  86. Feng S, Coward J, McCaffrey E, et al. Pembrolizumab-Induced Encephalopathy: A Review of Neurological Toxicities with Immune Checkpoint Inhibitors. J Thorac Oncol 2017; 12:1626.
  87. Spain L, Walls G, Julve M, et al. Neurotoxicity from immune-checkpoint inhibition in the treatment of melanoma: a single centre experience and review of the literature. Ann Oncol 2017; 28:377.
  88. Larkin J, Chmielowski B, Lao CD, et al. Neurologic Serious Adverse Events Associated with Nivolumab Plus Ipilimumab or Nivolumab Alone in Advanced Melanoma, Including a Case Series of Encephalitis. Oncologist 2017; 22:709.
  89. Dubey D, David WS, Reynolds KL, et al. Severe Neurological Toxicity of Immune Checkpoint Inhibitors: Growing Spectrum. Ann Neurol 2020; 87:659.
  90. Puzanov I, Diab A, Abdallah K, 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.
  91. Wilgenhof S, Neyns B. Anti-CTLA-4 antibody-induced Guillain-Barré syndrome in a melanoma patient. Ann Oncol 2011; 22:991.
  92. Eggermont AM, Chiarion-Sileni V, Grob JJ, et al. Ipilimumab versus placebo after complete resection of stage III melanoma: Initial efficacy and safety results from the EORTC 18071 phase III trial. J Clin Oncol 2014; 32S: ASCO #LBA9008.
  93. Safa H, Johnson DH, Trinh VA, et al. Immune checkpoint inhibitor related myasthenia gravis: single center experience and systematic review of the literature. J Immunother Cancer 2019; 7:319.
  94. Johnson DB, Manouchehri A, Haugh AM, et al. Neurologic toxicity associated with immune checkpoint inhibitors: a pharmacovigilance study. J Immunother Cancer 2019; 7:134.
  95. Maur M, Tomasello C, Frassoldati A, et al. Posterior reversible encephalopathy syndrome during ipilimumab therapy for malignant melanoma. J Clin Oncol 2012; 30:e76.
  96. Bot I, Blank CU, Boogerd W, Brandsma D. Neurological immune-related adverse events of ipilimumab. Pract Neurol 2013; 13:278.
  97. Nannini S, Koshenkova L, Baloglu S, et al. Immune-related aseptic meningitis and strategies to manage immune checkpoint inhibitor therapy: a systematic review. J Neurooncol 2022; 157:533.
  98. Bhatia S, Huber BR, Upton MP, Thompson JA. Inflammatory enteric neuropathy with severe constipation after ipilimumab treatment for melanoma: a case report. J Immunother 2009; 32:203.
  99. Liao B, Shroff S, Kamiya-Matsuoka C, Tummala S. Atypical neurological complications of ipilimumab therapy in patients with metastatic melanoma. Neuro Oncol 2014; 16:589.
  100. Vitt JR, Kreple C, Mahmood N, et al. Autoimmune pancerebellitis associated with pembrolizumab therapy. Neurology 2018; 91:91.
  101. Nersesjan V, McWilliam O, Krarup LH, Kondziella D. Autoimmune Encephalitis Related to Cancer Treatment With Immune Checkpoint Inhibitors: A Systematic Review. Neurology 2021; 97:e191.
  102. Reynolds KL, Guidon AC. Diagnosis and Management of Immune Checkpoint Inhibitor-Associated Neurologic Toxicity: Illustrative Case and Review of the Literature. Oncologist 2019; 24:435.
  103. Dolladille C, Akroun J, Morice PM, et al. Cardiovascular immunotoxicities associated with immune checkpoint inhibitors: a safety meta-analysis. Eur Heart J 2021; 42:4964.
  104. Laenens D, Yu Y, Santens B, et al. Incidence of Cardiovascular Events in Patients Treated With Immune Checkpoint Inhibitors. J Clin Oncol 2022; 40:3430.
  105. Johnson DB, Balko JM, Compton ML, et al. Fulminant Myocarditis with Combination Immune Checkpoint Blockade. N Engl J Med 2016; 375:1749.
  106. Naqash AR, Moey MYY, Cherie Tan XW, et al. Major Adverse Cardiac Events With Immune Checkpoint Inhibitors: A Pooled Analysis of Trials Sponsored by the National Cancer Institute-Cancer Therapy Evaluation Program. J Clin Oncol 2022; 40:3439.
  107. Salem JE, Allenbach Y, Vozy A, et al. Abatacept for Severe Immune Checkpoint Inhibitor-Associated Myocarditis. N Engl J Med 2019; 380:2377.
  108. Gordon IO, Wade T, Chin K, et al. Immune-mediated red cell aplasia after anti-CTLA-4 immunotherapy for metastatic melanoma. Cancer Immunol Immunother 2009; 58:1351.
  109. Akhtari M, Waller EK, Jaye DL, et al. Neutropenia in a patient treated with ipilimumab (anti-CTLA-4 antibody). J Immunother 2009; 32:322.
  110. Delyon J, Mateus C, Lambert T. Hemophilia A induced by ipilimumab. N Engl J Med 2011; 365:1747.
  111. Bulbul A, Mustafa A, Chouial S, Rashad S. Idiopathic thrombocytopenic purpura and autoimmune neutropenia induced by prolonged use of nivolumab in Hodgkin's lymphoma. Ann Oncol 2017; 28:1675.
  112. Pellegrino B, Musolino A, Tiseo M. Anti-PD-1-related cryoglobulinemia during treatment with nivolumab in NSCLC patient. Ann Oncol 2017.
  113. Helgadottir H, Kis L, Ljungman P, et al. Lethal aplastic anemia caused by dual immune checkpoint blockade in metastatic melanoma. Ann Oncol 2017; 28:1672.
  114. Shiuan E, Beckermann KE, Ozgun A, et al. Thrombocytopenia in patients with melanoma receiving immune checkpoint inhibitor therapy. J Immunother Cancer 2017; 5:8.
  115. Haddad TC, Zhao S, Li M, et al. Immune checkpoint inhibitor-related thrombocytopenia: incidence, risk factors and effect on survival. Cancer Immunol Immunother 2022; 71:1157.
  116. Summary Safety Review - Tecentriq (atezolizumab) - Health Canada https://hpr-rps.hres.ca/reg-content/summary-safety-review-detail.php?lang=en&linkID=SSR00258 (Accessed on February 09, 2021).
  117. Tanios GE, Doley PB, Munker R. Autoimmune hemolytic anemia associated with the use of immune checkpoint inhibitors for cancer: 68 cases from the Food and Drug Administration database and review. Eur J Haematol 2019; 102:157.
  118. Leaf RK, Ferreri C, Rangachari D, et al. Clinical and laboratory features of autoimmune hemolytic anemia associated with immune checkpoint inhibitors. Am J Hematol 2019; 94:563.
  119. Summary safety review: Nivolumab and ipilimumab used alone, or in combination. Health Canada. Government of Canada. https://hpr-rps.hres.ca/reg-content/summary-safety-review-detail.php?lang=en&linkID=SSR00225 (Accessed on July 11, 2019).
  120. Noseda R, Bertoli R, Müller L, Ceschi A. Haemophagocytic lymphohistiocytosis in patients treated with immune checkpoint inhibitors: analysis of WHO global database of individual case safety reports. J Immunother Cancer 2019; 7:117.
  121. Cappelli LC, Gutierrez AK, Baer AN, et al. Inflammatory arthritis and sicca syndrome induced by nivolumab and ipilimumab. Ann Rheum Dis 2017; 76:43.
  122. Cappelli LC, Gutierrez AK, Bingham CO 3rd, Shah AA. Rheumatic and Musculoskeletal Immune-Related Adverse Events Due to Immune Checkpoint Inhibitors: A Systematic Review of the Literature. Arthritis Care Res (Hoboken) 2017; 69:1751.
  123. Cappelli LC, Naidoo J, Bingham CO 3rd, Shah AA. Inflammatory arthritis due to immune checkpoint inhibitors: challenges in diagnosis and treatment. Immunotherapy 2017; 9:5.
  124. Daxini A, Cronin K, Sreih AG. Vasculitis associated with immune checkpoint inhibitors-a systematic review. Clin Rheumatol 2018; 37:2579.
  125. Patrinely JR Jr, Johnson R, Lawless AR, et al. Chronic Immune-Related Adverse Events Following Adjuvant Anti-PD-1 Therapy for High-risk Resected Melanoma. JAMA Oncol 2021; 7:744.
  126. Atezolizumab: Risk of immune-related myositis. Recalls and safety alerts. Government of Canada. http://healthycanadians.gc.ca/recall-alert-rappel-avis/hc-sc/2019/69274a-eng.php (Accessed on March 15, 2019).
  127. Abu-Sbeih H, Ali FS, Naqash AR, et al. Resumption of Immune Checkpoint Inhibitor Therapy After Immune-Mediated Colitis. J Clin Oncol 2019; 37:2738.
  128. Weber JS, Kudchadkar RR, Yu B, et al. Safety, efficacy, and biomarkers of nivolumab with vaccine in ipilimumab-refractory or -naive melanoma. J Clin Oncol 2013; 31:4311.
  129. Menzies AM, Johnson DB, Ramanujam S, et al. Anti-PD-1 therapy in patients with advanced melanoma and preexisting autoimmune disorders or major toxicity with ipilimumab. Ann Oncol 2017; 28:368.
  130. Pollack MH, Betof A, Dearden H, et al. Safety of resuming anti-PD-1 in patients with immune-related adverse events (irAEs) during combined anti-CTLA-4 and anti-PD1 in metastatic melanoma. Ann Oncol 2018; 29:250.
  131. Santini FC, Rizvi H, Plodkowski AJ, et al. Safety and Efficacy of Re-treating with Immunotherapy after Immune-Related Adverse Events in Patients with NSCLC. Cancer Immunol Res 2018; 6:1093.
  132. Simonaggio A, Michot JM, Voisin AL, et al. Evaluation of Readministration of Immune Checkpoint Inhibitors After Immune-Related Adverse Events in Patients With Cancer. JAMA Oncol 2019; 5:1310.
  133. Weber JS, Postow M, Lao CD, Schadendorf D. Management of Adverse Events Following Treatment With Anti-Programmed Death-1 Agents. Oncologist 2016; 21:1230.
  134. Li M, Sack JS, Rahma OE, et al. Outcomes after resumption of immune checkpoint inhibitor therapy after high-grade immune-mediated hepatitis. Cancer 2020; 126:5088.
  135. Arbour KC, Mezquita L, Long N, et al. Impact of Baseline Steroids on Efficacy of Programmed Cell Death-1 and Programmed Death-Ligand 1 Blockade in Patients With Non-Small-Cell Lung Cancer. J Clin Oncol 2018; 36:2872.
  136. Schadendorf D, Ascierto PA, Haanen J, et al. Safety and efficacy of nivolumab in challenging subgroups with advanced melanoma who progressed on or after ipilimumab treatment: A single-arm, open-label, phase II study (CheckMate 172). Eur J Cancer 2019; 121:144.
  137. Abdel-Wahab N, Shah M, Lopez-Olivo MA, Suarez-Almazor ME. Use of Immune Checkpoint Inhibitors in the Treatment of Patients With Cancer and Preexisting Autoimmune Disease: A Systematic Review. Ann Intern Med 2018; 168:121.
  138. Abu-Sbeih H, Faleck DM, Ricciuti B, et al. Immune Checkpoint Inhibitor Therapy in Patients With Preexisting Inflammatory Bowel Disease. J Clin Oncol 2020; 38:576.
  139. Johnson DB, Sullivan RJ, Ott PA, et al. Ipilimumab Therapy in Patients With Advanced Melanoma and Preexisting Autoimmune Disorders. JAMA Oncol 2016; 2:234.
  140. Tison A, Quéré G, Misery L, et al. Safety and Efficacy of Immune Checkpoint Inhibitors in Patients With Cancer and Preexisting Autoimmune Disease: A Nationwide, Multicenter Cohort Study. Arthritis Rheumatol 2019; 71:2100.
  141. Grover S, Ruan AB, Srivoleti P, et al. Safety of Immune Checkpoint Inhibitors in Patients With Pre-Existing Inflammatory Bowel Disease and Microscopic Colitis. JCO Oncol Pract 2020; 16:e933.
  142. Danlos FX, Voisin AL, Dyevre V, et al. Safety and efficacy of anti-programmed death 1 antibodies in patients with cancer and pre-existing autoimmune or inflammatory disease. Eur J Cancer 2018; 91:21.
  143. Tang K, Tiu BC, Wan G, et al. Pre-Existing Autoimmune Disease and Mortality in Patients Treated with Anti-PD-1 and Anti-PD-L1 Therapy. J Natl Cancer Inst 2022; 114:1200.
  144. Han CY, Fitzgerald C, Lee M, et al. Association Between Toxic Effects and Survival in Patients With Cancer and Autoimmune Disease Treated With Checkpoint Inhibitor Immunotherapy. JAMA Oncol 2022; 8:1352.
  145. Lee C, Drobni ZD, Zafar A, et al. Pre-Existing Autoimmune Disease Increases the Risk of Cardiovascular and Noncardiovascular Events After Immunotherapy. JACC CardioOncol 2022; 4:660.
  146. Halle BR, Betof Warner A, Zaman FY, et al. Immune checkpoint inhibitors in patients with pre-existing psoriasis: safety and efficacy. J Immunother Cancer 2021; 9.
  147. Haanen J, Ernstoff MS, Wang Y, et al. Autoimmune diseases and immune-checkpoint inhibitors for cancer therapy: review of the literature and personalized risk-based prevention strategy. Ann Oncol 2020; 31:724.
  148. Ramnaraign BH, Chatzkel JA, Al-Mansour ZA, et al. Immunotherapy Management in Special Cancer Patient Populations. JCO Oncol Pract 2021; 17:240.
  149. Elias R, Giobbie-Hurder A, McCleary NJ, et al. Efficacy of PD-1 & PD-L1 inhibitors in older adults: a meta-analysis. J Immunother Cancer 2018; 6:26.
  150. Nishijima TF, Shachar SS, Nyrop KA, Muss HB. Safety and Tolerability of PD-1/PD-L1 Inhibitors Compared with Chemotherapy in Patients with Advanced Cancer: A Meta-Analysis. Oncologist 2017; 22:470.
  151. Friedman CF, Wolchok JD. Checkpoint inhibition and melanoma: Considerations in treating the older adult. J Geriatr Oncol 2017; 8:237.
  152. Nebhan CA, Cortellini A, Ma W, et al. Clinical Outcomes and Toxic Effects of Single-Agent Immune Checkpoint Inhibitors Among Patients Aged 80 Years or Older With Cancer: A Multicenter International Cohort Study. JAMA Oncol 2021; 7:1856.
  153. Wong SK, Blum SM, Sun X, et al. Efficacy and safety of immune checkpoint inhibitors in young adults with metastatic melanoma. Eur J Cancer 2023; 181:188.
Topic 96368 Version 106.0

References

آیا می خواهید مدیلیب را به صفحه اصلی خود اضافه کنید؟