The emergence of immune checkpoint inhibitors for solid tumor treatments represents a major oncologic advance. Since the approval of ipilimumab, a cytotoxic T-lymphocyte–associated antigen 4 (CTLA-4) antibody, for the treatment of metastatic melanoma, many drugs, especially those targeting PD-1/PD-L1, have demonstrated promising antitumor effects in many types of cancer. By reactivating the immune system, these immunotherapies have led to the development of new toxicity profiles, also called immune-related adverse events (irAE). IrAEs can involve many organ systems, and their management is radically different from that of cytotoxic drugs; irAEs require immunosuppressive treatments, such as corticoids or TNFα antibody. In addition, the occurrence of irAEs has raised significant questions. Here, we summarize progress that has been made toward answering these questions, focusing on (i) the impact of immunotherapy dose on irAE occurrence, (ii) the correlation between irAE and patient outcome, (iii) the safety of immune checkpoint inhibitors in patients already treated for autoimmune disease, and (iv) the suspected effect on tumor growth of steroids used for the management of irAEs. Clin Cancer Res; 22(18); 4550–5. ©2016 AACR.

Immunotherapies soaring

Immunotherapy has recently led to a paradigm shift in cancer treatment, in which the main target is the immune system instead of the cancer cells. Indeed, cancer cells are known to bypass host immunosurveillance (i.e., the ability of the immune system to recognize and destroy cancer cells), leading to tumor growth (1). One mechanism by which cancer cells escape the immune system is by overexpressing immunosuppressive surface ligands that interact with T-cell molecules, such as cytotoxic T-lymphocyte–associated antigen 4 (CTLA-4) or programmed cell death protein (PD-1), leading to T-cell exhaustion (Fig. 1; ref. 2). This knowledge led to the development of immune checkpoint inhibitors, which can antagonize these immune inhibitory pathways. The anti–CTLA-4 monoclonal antibody ipilimumab was an early inhibitor to appear in this area and has been approved for the treatment of metastatic melanoma (3). Since then, many new agents, especially those targeting the PD-1/programmed cell death ligand 1 (PD-1/PD-L1) complex, such as the anti–PD-1 antibodies pembrolizumab and nivolumab, or the PD-L1 antibodies atezolizumab and durvalumab, have demonstrated promising effects in many different tumor types (4–10). In addition, pembrolizumab and nivolumab have recently been approved for the treatment of metastatic melanoma and non–small cell lung cancer (NSCLC) patients. Another escape mechanism involves tumor-associated macrophages (TAM; Fig. 1) and the colony-stimulating factor-1 receptor (CSF-1R). CSF-1R is a tyrosine kinase receptor involved in the differentiation and survival of macrophages and their precursors. TAMs can suppress antitumor immunity and have been associated with poorer patient outcomes (11). On the basis of these data, CSF-1R antibodies are under clinical development (12). By reactivating the IS, these immunomodulatory antibodies have led to the emergence of unusual autoimmune toxicities, also called immune-related adverse events (irAE; ref. 13). IrAEs management is challenging because they may concern many organ systems, including the skin, hepatic, gastrointestinal, endocrine, and pulmonary systems (14). As irAEs often require steroid treatments, which are thought to affect antitumor treatment efficacy, physicians require training to manage these irAEs and achieve the best patient care.

Figure 1.

Targeting tumor microenvironment with immune checkpoint antibodies. A, tumor microenvironment representation. CD8+ T-cell activation requires antigen presentation (peptide) by a major histocompatibility complex (MHC) molecule in addition to the costimulatory signal (B7 interaction on an antigen-presenting cell with CD28). However, CTLA-4 upregulation after T-cell activation downregulates T-cell immune function. PD-1 is a negative regulator of T-cell function. When PD-1 interacts with its ligand, PD-L1, the tumor cells can escape immunosurveillance. B, CSF-1R and its ligand CSF-1 regulate the function of TAMs, involved in tumorigenesis. Anti–CSF-1R or anti–CSF-1 antibodies are under investigation to block CSF-1R signaling.

Figure 1.

Targeting tumor microenvironment with immune checkpoint antibodies. A, tumor microenvironment representation. CD8+ T-cell activation requires antigen presentation (peptide) by a major histocompatibility complex (MHC) molecule in addition to the costimulatory signal (B7 interaction on an antigen-presenting cell with CD28). However, CTLA-4 upregulation after T-cell activation downregulates T-cell immune function. PD-1 is a negative regulator of T-cell function. When PD-1 interacts with its ligand, PD-L1, the tumor cells can escape immunosurveillance. B, CSF-1R and its ligand CSF-1 regulate the function of TAMs, involved in tumorigenesis. Anti–CSF-1R or anti–CSF-1 antibodies are under investigation to block CSF-1R signaling.

Close modal

General safety profiles

IrAEs affect generally the tracts involved in common autoimmune diseases. They are here reported according to the NCI-CTCAE version 4.0. Anti–CTLA-4 antibodies mostly affect the skin and the gastrointestinal tract, as observed in 44% and 35% of cases, respectively, whereas the endocrine (6%) and hepatic (5%) systems are rarely affected (15). Skin adverse events (AE) are usually the first to appear (e.g., after 3 weeks), whereas gastrointestinal AEs can be expected at approximately 5 to 10 weeks, and liver AEs and hypophysitis appear later. Unlike endocrine events, most AEs can be reversed within a few weeks (16). Although AEs are frequent, they are usually low grade; low-grade AEs were observed in 85% of patients treated with ipilimumab for metastatic melanoma, but grade 3 to 4 AEs were observed in 24% of patients, mainly in the gastrointestinal tract (11%; ref. 15). The most common skin AEs are papular rashes associated with pruritus (skin itching), whereas alopecia (hair loss) and vitiligo (skin hypopigmentation) are rarely described. Being reported in 13% of clinical trials, hypophysitis is the most frequent endocrine AE. Hypo- or hyperthyroidism are very uncommon (<6%). Colitis, indicated by diarrhea, abdominal pain and rectal bleeding, is the main gastrointestinal AE and the most severe irAE associated with anti–CTLA-4 (15). Although rare, cases of fatal bowel perforation have been described (<0.5%; ref. 17). Finally, the death rate due to CTLA-4 antibodies (ipilimumab or tremelimumab) is less than 1% (11/1,265 patients; ref.15). Few data exist about long-term toxicities related to ipilimumab. Most of the chronic side effects concern endocrine toxicities with the need of long-term hormone replacement. Chronic neurologic complications raise questions about the role of whole-brain radiotherapy in patients treated with ipilimumab (18).

The AE profiles of PD-1 or PD-L1 antibodies are very similar; however, the side effects of PD-1 and PD-L1 antibodies are less frequent and less severe than those of CTLA-4 antibodies. The main AE is asthenia (16%–34%), followed by decreased appetite (5%–19%), rash (16%), diarrhea (14%), and nausea (15%), which are mainly low grade. High-grade AEs occur in only 7% to 12% of patients (7, 19–23). Interestingly, those AEs considered irAEs are very uncommon and arise later; these include hypothyroidism (8%), hyperthyroidism (5%), and pneumonitis (4%), and very rarely, colitis or hepatitis. Deaths related to the treatment rate accounted for less than 1%, mostly due to pneumonitis (7, 19–22).

The AE profile of CSF-1R inhibitors is well characterized. Whether the disease is benign (e.g., pigmented villonodular synovitis) or malignant (e.g., solid tumors), asthenia affects 56% to 70% of patients; facial edema, pruritus, rash, and peripheral edema affect 64%, 56%, 40%, and up to 44% of patients, respectively (24, 25). Interestingly, autoimmune disorders such as lupus have been described with such agents but only in patients with a benign condition (pigmented villonodular synovitis) suggesting a role of the patient's immune status in their occurrence (24).

Rare adverse events

As immune checkpoint inhibitors have recently entered clinical practice, they are thought to be involved in many rare AEs, the most relevant of which are briefly described here.

Several neurologic events have been observed in patients treated with ipilimumab, including Guillain-Barré syndrome, encephalopathy, aseptic meningitis, myasthenia (characterized by muscle weakness), and myopathy. Three deaths due to neurologic AEs have been linked to ipilimumab (26–30). Other rare side effects are mostly controlled with appropriate steroid-based treatments. Several hematologic side effects have also been attributed to ipilimumab, including one case each of neutropenia, thrombopenia, red cell aplasia, and hemophilia A (31–34). Furthermore, ipilimumab has been associated with several ocular effects, including orbital inflammation, uveitis, and keratitis (35). Other rare side effects include lupus nephritis (36), giant cell arteritis (37), and more recently, a case of toxic epidermal necrolysis-like reaction requiring cyclosporine treatment (38).

Most rare AEs due to PD-1/PD-L1 antibodies are manageable with dedicated treatment. One death was attributable to multifocal central nervous system demyelination occurring under nivolumab treatment (39). Two case reports have described nivolumab involvement in myasthenia occurring in melanoma patients: In one case, nivolumab was associated with ipilimumab treatment; in the other case, both myasthenia and rhabdomyolysis occurred when nivolumab was used alone (40, 41). In addition, one case of myositis occurred during nivolumab treatment, and the main symptom was respiratory discomfort (42). Regarding skin AEs, nivolumab treatment has been associated with several cases of psoriasis exacerbation, all occurring in patients with melanoma (43, 44). Lichenoid dermatitis occurred in 3 patients treated with pembrolizumab for melanoma (45). Pembrolizumab has also been associated with one case of acute autoimmune myocarditis. Fortunately, the symptoms rapidly improved after appropriate corticosteroid and heart-failure treatments (46). Cases of pancreatitis, retinal vasculitis, fasciitis, and arthritis have also been attributed to pembrolizumab treatment (47–50). Recently, 3 cases of sarcoidosis in sarcoma patients receiving pembrolizumab treatment have been described (51). Similar cases were previously reported in melanoma patients treated with ipilimumab (52–54). Although these patients were mostly asymptomatic, the main indicator was enlarged mediastinal lymph nodes visible on CT scans, which could be confounded with tumor progression.

Combination toxicities: immune checkpoint inhibitors with or without tyrosine kinase inhibitors

The combination of ipilimumab and nivolumab was recently approved for treating advanced or metastatic melanoma (21). Compared with monotherapies, combination treatments [nivolumab, 1 mg/kg, plus ipilimumab, 3 mg/kg every 3 weeks for 4 doses] needed to be discontinued more frequently, with rates of 36.4% versus 14.8% and 7.7% for ipilimumab and nivolumab combined and for each drug individually, respectively. Discontinuation was mostly due to diarrhea and colitis. Grade 3–4 side effects were more frequent than in monotherapy arms (nivolumab, 3 mg/kg every 2 weeks, or ipilimumab, 3 mg/kg every 3 weeks): 55% versus 16.3% and 27.3% respectively. No deaths were related to the combination. On the contrary, in NSCLC patients, the association of nivolumab, 3 mg/kg, plus ipilimumab, 3 mg/kg, was responsible for 2 deaths: One was due to myasthenia gravis and one due to renal failure. In the group, nivolumab, 3 mg/mg, plus ipilimumab, 1 mg/mg, every 3 weeks, one death was due to pneumonitis related to the treatment (55).

Immune checkpoint and tyrosine kinase inhibitor combinations are now being tested. Combinations of nivolumab plus sunitinib (N+S) or pazopanib (N+P) were evaluated in patients with metastatic renal cell carcinoma. One hundred percent of patients had at least one AE, irrespective of the treatment. Hypertension was more frequent with (N+S) than with (N+P), at rates of 48% and 25%, respectively. Similar results were observed for treatment discontinuation, at 36.4% and 25%, respectively, mostly due to increased alanine or aspartate aminotransferase. Other combinations are currently being evaluated, including bevacizumab ± MPDL3280A (NCT01984242), pazopanib + pembrolizumab (NCT02014636), and axitinib + pembrolizumab (NCT02133742; ref. 56).

A dose effect?

As several irAEs occur after long delays, determining the most effective immunotherapy dose associated with the least toxicity is challenging.

Three studies have examined irAE occurrence versus ipilimumab dose. When comparing 10 mg/kg to 3 mg/kg, the risk ratios of global irAE incidence were not different [RR, 1.16; 95% confidence interval (CI) 0.97–1.38]. The difference was only statistically significant for high-grade (i.e., grade 3–4) irAEs (RR, 3.1; 95% CI, 1.59–6.03), which affect mostly the gastrointestinal tract (15). Using whole-blood gene-expression profiling in melanoma patients treated with ipilimumab, Shahabi and colleagues (57) found that the neutrophil-activation markers CD177 and CEACAM1 were associated with gastrointestinal irAE occurrence. These data suggest a possible role of neutrophils in ipilimumab-associated gastrointestinal irAEs.

There are no data about the potential dose effects of PD-1/PD-L1 antibodies on toxicity. Only one study has suggested that there are no AE profile differences between NSCLC patients treated with different doses of pembrolizumab, i.e., 10 mg/kg every 2 or 3 weeks (58).

Correlation between irAE occurrence and outcome

Data are inconsistent about the impact of irAE occurrence on the outcomes of melanoma patients treated with ipilimumab. In 56 patients treated with both ipilimumab and gp100 melanoma-associated antigen, Attia and colleagues (59) showed a significant improvement in the overall response rate (ORR) of patients who experienced high-grade irAEs, at 36% versus 5% (P = 0.008). Some case series have brought better ORRs in renal cell carcinoma and melanoma patients treated with ipilimumab who developed enterocolitis or hypophysitis than in those without AEs (60, 61). However, irAEs were noticed in 254 out of 298 patients (85%) treated with a standard dose of ipilimumab. When comparing the outcomes of patients without any irAEs and those of patients with irAEs of any grade, no differences in time to treatment failure and overall survival were observed (62). Whether irAE severity can be used as a predictive factor of treatment efficacy in patients treated with ipilimumab remains under debate.

Skin irAEs seems to be correlated with outcome in melanoma patients treated with PD-1 antibodies. Vitiligo has been described in 11% of melanoma patients treated with pembrolizumab (63). Interestingly, this AE has recently been associated with favorable outcomes. In 67 melanoma patients treated with pembrolizumab, 17 patients (25%) developed vitiligo, and the ORRs were 71% and 28% in patients with and without vitiligo, respectively (64). Another study evaluated the incidence of skin AEs and outcome. Eighty-three patients were treated for lung cancer, melanoma, or prostate cancer with different schedules of pembrolizumab (10 mg/kg every 3 weeks; 10 mg/kg every 2 weeks; 2 mg/kg/3 per week). Thirty-five (42%) developed cutaneous irAEs, with the most frequent being maculopapular eruption, pruritus, and hypopigmentation. Regardless of pembrolizumab schedule, time to progression was significantly better in patients who experienced skin irAEs than in those who did not (65). Similarly, in patients (n = 112) treated with nivolumab for advanced or metastatic melanoma, vitiligo and rash were associated with a significant improvement in OS: HR, 0.22; 95% CI, 0.03–0.81 and HR, 0.45; 95% CI, 0.25–0.77, respectively (66). Taken together, these data suggest a predictive role of skin irAE occurrence in patients receiving a PD-1 inhibitor.

Is it dangerous to treat patients having underlying autoimmune disease or chronic immunosuppression with immune checkpoint inhibitors?

By antagonizing immune inhibitory pathways, immune checkpoint inhibitor treatments were thought to potentially worsen autoimmune disease.

A report on the largest group of patients with autoimmune disease who underwent ipilimumab treatment for metastatic melanoma was recently published (67). The autoimmune diseases included rheumatoid arthritis, psoriasis, inflammatory bowel disease, systemic lupus erythematosus, multiple sclerosis, and autoimmune thyroiditis. Of the 30 patients, 50% experienced neither an irAE nor autoimmune disease exacerbation. Twenty-seven percent experienced autoimmune disease exacerbation, and 33% experienced high-grade irAEs. One death was associated with immune-related colitis. Few case reports have contributed other data; ipilimumab treatment appeared to be safe, especially in 2 kidney transplant patients, and only one patient with ulcerative colitis experienced autoimmune disease exacerbation during ipilimumab treatment (68–70).

No data are currently available about the effects of pembrolizumab treatment in patients with autoimmune disease. Two case reports have described the safety profile of pembrolizumab in patients with HCV alone or with HCV/HIV coinfection (71). Few data have shown thyroiditis aggravation in patients treated with nivolumab (72). Recently, a case report highlighted renal allograft rejection after 2 months of pembrolizumab treatment in a patient treated for metastatic cutaneous squamous cell carcinoma (73).

Of note, most of the patients in these cases were, at the time of the checkpoint inhibitor treatment, actively treated for their autoimmune or viral disease (by corticoid or immunosuppressive agents such as tacrolimus, mycophenolate, or antiviral therapies). The paucity of these data supports the necessity of additional investigations of checkpoint inhibitors in patients with immunodeficiencies or autoimmune disease. Currently, immune checkpoint inhibitors should be used with caution in these patients.

Does corticosteroid use really decrease the efficacy of immune checkpoint inhibitors?

Although steroids are thought to potentially reverse the antitumor effects of immunotherapies via their immunosuppressive effect, no data support this idea. In a pooled analysis of 139 metastatic melanoma patients treated with CTLA-4 blockade in a phase II trial, irAE management via high-dose steroids did not affect ORR (74). Of 298 melanoma patients treated with ipilimumab included in a study by Yang and colleagues, 103 patients (35%) required systemic corticosteroid (mostly 1 week or longer) treatments for irAEs. Twenty-nine (10%) patients required anti-TNFα therapy. No differences in OS and TTF were found (60) between patients who did and did not receive steroids. The impact of anti-TNFα on outcomes was not evaluable in this study with regard to the few patients who received this treatment. However, recent data have shown no worse outcomes in patients with diarrhea related to ipilimumab treated with infliximab (n = 7) compared with the other drug (n = 12; ref. 75). Similar results concerning the use of steroids were documented in two case reports of severe irAEs in melanoma treated with ipilimumab; corticoid treatments did not affect outcomes (76). No data are available about PD-1/PD-L1 or CSF-1R inhibitors in this context. However, regarding the effect of checkpoint inhibitors on the immune system, corticosteroids and other immunosuppressive treatments clearly need to be used according to their recommendations to treat irAEs (77). Guidelines recommend a short course of oral or i.v. steroids, at a dose of 1 to 4 mg/kg, associated with oral antihistamines in the presence of pruritus, with a rapid indication for treatment with infliximab or other immunosuppressive therapies if there is no improvement after 48 to 72 hours or if symptoms worsen (78).

Immune checkpoint inhibition is typically associated with transient irAEs, the majority of which resolve within a few weeks, excluding endocrine side effects. The rapid identification of these irAEs and the initiation of systemic immunosuppression can improve patients' outcomes without compromising antitumor treatment efficacy. The development and implementation of immune checkpoint inhibitors, along with the presence of delayed toxicities, have raised questions and altered conventional anticancer drug development; these treatments have more of an on/off effect (79) than a dose effect, and no MTD has been defined. There is no rationale for dose repetition (58), and the optimal treatment duration is unknown because long responders have been observed, raising the question of whether to stop treatment or administer maintenance treatments (80). More important than an MTD is, the challenge for these new immunotherapies to identify minimum active doses, thereby achieving better tumor control and minimizing irAEs for optimal patient care, especially now with the arrival of the checkpoint inhibitors in earlier settings than metastatic disease (81).

No potential conflicts of interest were disclosed.

Conception and design: A. Italiano

Development of methodology: S. Cousin, A. Italiano

Acquisition of data (provided animals, acquired and managed patients, provided facilities, etc.): A. Italiano

Analysis and interpretation of data (e.g., statistical analysis, biostatistics, computational analysis): S. Cousin, A. Italiano

Writing, review, and/or revision of the manuscript: S. Cousin, A. Italiano

Administrative, technical, or material support (i.e., reporting or organizing data, constructing databases): A. Italiano

Study supervision: A. Italiano

1.
Schreiber
RD
,
Old
LJ
,
Smyth
MJ
. 
Cancer immunoediting: integrating immunity's roles in cancer suppression and promotion
.
Science
2011
;
331
:
1565 70
.
2.
Mellman
I
,
Coukos
G
,
Dranoff
G
. 
Cancer immunotherapy comes of age
.
Nature
2011
;
480
:
480
9
.
3.
Hodi
FS
,
O'Day
SJ
,
McDermott
DF
,
Weber
RW
,
Sosman
JA
,
Haanen
JB
, et al
Improved survival with ipilimumab in patients with metastatic melanoma
.
N Engl J Med
2010
;
363
:
711
23
.
4.
Weber
JS
,
D'Angelo
SP
,
Minor
D
,
Hodi
FS
,
Gutzmer
R
,
Neyns
B
, et al
Nivolumab versus chemotherapy in patients with advanced melanoma who progressed after anti–CTLA-4 treatment (CheckMate 037): a randomised, controlled, open-label, phase 3 trial
.
Lancet Oncol
2015
;
16
:
375
84
.
5.
Herbst
RS
,
Baas
P
,
Kim
D
,
Felip
E
,
Pérez-Gracia
JL
,
Han
J
, et al
Pembrolizumab versus docetaxel for previously treated, PD-L1-positive, advanced non–small cell lung cancer (KEYNOTE-010): a randomised controlled trial
.
Lancet
2016
;
387
:
1540
50
.
6.
Borghaei
H
,
Paz-Ares
L
,
Horn
L
,
Spigel
DR
,
Steins
M
,
Ready
NE
, et al
Nivolumab versus docetaxel in advanced nonsquamous non–small cell Lung cancer
.
N Engl J Med
2015
;
373
:
1627
39
.
7.
Brahmer
J
,
Reckamp
KL
,
Baas
P
,
Crinò
L
,
Eberhardt
WE
,
Poddubskaya
E
, et al
Nivolumab versus docetaxel in advanced squamous-cell non–small cell Lung cancer
.
N Engl J Med
2015
;
373
:
123
35
.
8.
Powles
T
,
Eder
JP
,
Fine
GD
,
Braiteh
FS
,
Loriot
Y
,
Cruz
C
, et al
MPDL3280A (anti–PD-L1) treatment leads to clinical activity in metastatic bladder cancer
.
Nature
2014
;
515
:
558
62
.
9.
Fehrenbacher
L
,
Spira
A
,
Ballinger
M
,
Kowanetz
M
,
Vansteenkiste
J
,
Mazieres
J
, et al
Atezolizumab versus docetaxel for patients with previously treated non–small cell lung cancer (POPLAR): a multicentre, open-label, phase 2 randomised controlled trial
.
Lancet
2016
;
387:1837–46
.
10.
Rosenberg
JE
,
Hoffman-Censits
J
,
Powles
T
,
van der Heijden
MS
,
Balar
AV
,
Necchi
A
, et al
Atezolizumab in patients with locally advanced and metastatic urothelial carcinoma who have progressed following treatment with platinum-based chemotherapy: a single-arm, multicentre, phase 2 trial
.
Lancet
2016
;
387
:
1298
304
.
11.
Zhang
QW
,
Liu
L
,
Gong
CY
,
Shi
HS
,
Zeng
YH
,
Wang
XZ
, et al
Prognostic significance of tumor-associated macrophages in solid tumor: a meta-analysis of the literature
.
PLoS ONE
2012
;
7
:
e50946
.
12.
Ries
CH
,
Cannarile
MA
,
Hoves
S
,
Benz
J
,
Wartha
K
,
Runza
V
, et al
Targeting tumor-associated macrophages with anti-CSF-1R antibody reveals a strategy for cancer therapy
.
Cancer Cell
2014
;
25
:
846
59
.
13.
Devaud
C
,
John
LB
,
Westwood
JA
,
Darcy
PK
,
Kershaw
MH
. 
Immune modulation of the tumor microenvironment for enhancing cancer immunotherapy
.
Oncoimmunology
2013
;
2
:
e25961
.
14.
Michot
JM
,
Bigenwald
C
,
Champiat
S
,
Collins
M
,
Carbonnel
F
,
Postel-Vinay
S
, et al
Immune-related adverse events with immune checkpoint blockade: a comprehensive review
.
Eur J Cancer
2016
;
54
:
139
48
.
15.
Bertrand
A
,
Kostine
M
,
Barnetche
T
,
Truchetet
M-E
,
Schaeverbeke
T
. 
Immune related adverse events associated with anti-CTLA-4 antibodies: systematic review and meta-analysis
.
BMC Med
2015
;
13
:
211
.
16.
Weber
JS
,
Kähler
KC
,
Hauschild
A
. 
Management of immune-related adverse events and kinetics of response with ipilimumab
.
J Clin Oncol
2012
;
30
:
2691
7
.
17.
Mitchell
KA
,
Kluger
H
,
Sznol
M
,
Hartman
DJ
. 
Ipilimumab-induced perforating colitis
.
J Clin Gastroenterol
2013
;
47
:
781
5
.
18.
Johnson
DB
,
Friedman
DL
,
Berry
E
,
Decker
I
,
Ye
F
,
Zhao
S
, et al
Survivorship in immune therapy: assessing chronic immune toxicities, health outcomes, and functional status among long-term ipilimumab survivors at a single referral center
.
Cancer Immunol Res
2015
;
3
:
464
9
.
19.
Borghaei
H
,
Paz-Ares
L
,
Horn
L
,
Spigel
DR
,
Steins
M
,
Ready
NE
, et al
Nivolumab versus Docetaxel in advanced nonsquamous non–small cell Lung cancer
.
N Engl J Med
2015
;
373
:
1627
39
.
20.
Motzer
RJ
,
Escudier
B
,
McDermott
DF
,
George
S
,
Hammers
HJ
,
Srinivas
S
, et al
Nivolumab versus Everolimus in advanced renal-cell carcinoma
.
N Engl J Med
2015
;
373
:
1803
13
.
21.
Larkin
J
,
Chiarion-Sileni
V
,
Gonzalez
R
,
Grob
JJ
,
Cowey
CL
,
Lao
CD
, et al
Combined nivolumab and ipilimumab or monotherapy in untreated melanoma
.
N Engl J Med
2015
;
373
:
23
34
.
22.
Weber
JS
,
D'Angelo
SP
,
Minor
D
,
Hodi
FS
,
Gutzmer
R
,
Neyns
B
, et al
Nivolumab versus chemotherapy in patients with advanced melanoma who progressed after anti–CTLA-4 treatment (CheckMate 037): a randomised, controlled, open-label, phase 3 trial
.
Lancet Oncol
2015
;
16
:
375
84
.
23.
Weber
JS
,
Antonia
SJ
,
Topalian
SL
,
Schadendorf
D
,
Larkin
J
,
Sznol
M
, et al
Safety profile of nivolumab (NIVO) in patients (pts) with advanced melanoma (MEL): a pooled analysis
.
J Clin Oncol
33
, 
2015
(
suppl; abstr 9018
).
24.
Cassier
PA
,
Italiano
A
,
Gomez-Roca
CA
,
Le Tourneau
C
,
Toulmonde
M
,
Cannarile
MA
, et al
CSF1R inhibition with emactuzumab in locally advanced diffuse-type tenosynovial giant cell tumours of the soft tissue: a dose-escalation and dose-expansion phase 1 study
.
Lancet Oncol
2015
;
16
:
949
56
.
25.
Gomez-Roca
CA
,
Cassier
PA
,
Italiano
A
,
Cannarile
M
,
Ries
C
,
Brillouet
A
, et al
Phase I study of RG7155, a novel anti–CSF1R antibody, in patients with advanced/metastatic solid tumors
.
J Clin Oncol
33
, 
2015
(
suppl; abstr 3005
).
26.
Wilgenhof
S
,
Neyns
B
. 
Anti–CTLA-4 antibody-induced Guillain-Barré syndrome in a melanoma patient
.
Ann Oncol
2011
;
22
:
991
3
.
27.
Gaudy-Marqueste
C
,
Monestier
S
,
Franques
J
,
Cantais
E
,
Richard
M-A
,
Grob
J-J
. 
A severe case of ipilimumab-induced Guillain-Barré syndrome revealed by an occlusive enteric neuropathy: a differential diagnosis for ipilimumab-induced colitis
.
J Immunother
2013
;
36
:
77
8
.
28.
Conry
RM
,
Sullivan
JC
,
Nabors
LB
. 
Ipilimumab-induced encephalopathy with a reversible splenial lesion
.
Cancer Immunol Res
2015
;
3
:
598
601
.
29.
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
93
.
30.
Hunter
G
,
Voll
C
,
Robinson
CA
. 
Autoimmune inflammatory myopathy after treatment with ipilimumab
.
Can J Neurol Sci
2009
;
36
:
518
20
.
31.
Akhtari
M
,
Waller
EK
,
Jaye
DL
,
Lawson
DH
,
Ibrahim
R
,
Papadopoulos
NE
, et al
Neutropenia in a patient treated with ipilimumab (anti–CTLA-4 antibody)
.
J Immunother
2009
;
32
:
322
4
.
32.
Delyon
J
,
Mateus
C
,
Lambert
T
. 
Hemophilia A induced by ipilimumab
.
N Engl J Med
2011
;
365
:
1747
8
.
33.
Ahmad
S
,
Lewis
M
,
Corrie
P
,
Iddawela
M
. 
Ipilimumab-induced thrombocytopenia in a patient with metastatic melanoma
.
J Oncol Pharm Pract
2012
;
18
:
287
92
.
34.
Gordon
IO
,
Wade
T
,
Chin
K
,
Dickstein
J
,
Gajewski
TF
. 
Immune-mediated red cell aplasia after anti–CTLA-4 immunotherapy for metastatic melanoma
.
Cancer Immunol Immunother
2009
;
58
:
1351
3
.
35.
Papavasileiou
E
,
Prasad
S
,
Freitag
SK
,
Sobrin
L
,
Lobo
A
. 
Ipilimumab-induced ocular and orbital inflammation-A case series and review of the literature
.
Ocul Immunol Inflamm
2015
;24:
1
7
.
36.
Fadel
F
,
El Karoui
KE
,
Knebelmann
B
. 
Anti-CTLA4 antibody-induced lupus nephritis
.
N Engl J Med
2009
;
361
:
211
2
.
37.
Goldstein
BL
,
Gedmintas
L
,
Todd
DJ
. 
Drug-associated polymyalgia rheumatica/giant cell arteritis occurring in two patients after treatment with ipilimumab, an antagonist of ctla-4
.
Arthritis Rheumatol
2014
;
66
:
768
9
.
38.
Nayar
N
,
Briscoe
K
,
Fernandez Penas
P
. 
Toxic epidermal necrolysis-like reaction with severe satellite cell necrosis associated with nivolumab in a patient with Ipilimumab refractory metastatic melanoma
.
J Immunother
2016
;
39
:
149
52
.
39.
Maurice
C
,
Schneider
R
,
Kiehl
T-R
,
Bavi
P
,
Roehrl
MH
,
Mason
WP
, et al
Subacute CNS demyelination after treatment with nivolumab for melanoma
.
Cancer Immunol Res
2015
;
3
:
1299
302
.
40.
Loochtan
AI
,
Nickolich
MS
,
Hobson-Webb
LD
. 
Myasthenia gravis associated with ipilimumab and nivolumab in the treatment of small-cell lung cancer
.
Muscle Nerve
2015
;
52
:
307
8
.
41.
Shirai
T
,
Sano
T
,
Kamijo
F
,
Saito
N
,
Miyake
T
,
Kodaira
M
, et al
Acetylcholine receptor binding antibody-associated myasthenia gravis and rhabdomyolysis induced by nivolumab in a patient with melanoma
.
Jpn J Clin Oncol
2016
;
46
:
86
8
.
42.
Yoshioka
M
,
Kambe
N
,
Yamamoto
Y
,
Suehiro
K
,
Matsue
H
. 
Case of respiratory discomfort due to myositis after administration of nivolumab
.
J Dermatol
2015
;
42
:
1008
9
.
43.
Kato
Y
,
Otsuka
A
,
Miyachi
Y
,
Kabashima
K
. 
Exacerbation of psoriasis vulgaris during nivolumab for oral mucosal melanoma
.
J Eur Acad Dermatol Venereol
. 
2015
Sep 21
[Epub ahead of print]
.
44.
Matsumura
N
,
Ohtsuka
M
,
Kikuchi
N
,
Yamamoto
T
. 
Exacerbation of psoriasis during Nivolumab therapy for metastatic melanoma
.
Acta Derm Venereol
2016
;
96
:
259
60
.
45.
Joseph
RW
,
Cappel
M
,
Goedjen
B
,
Gordon
M
,
Kirsch
B
,
Gilstrap
C
, et al
Lichenoid dermatitis in three patients with metastatic melanoma treated with anti-PD-1 therapy
.
Cancer Immunol Res
2015
;
3
:
18
22
.
46.
Läubli
H
,
Balmelli
C
,
Bossard
M
,
Pfister
O
,
Glatz
K
,
Zippelius
A
. 
Acute heart failure due to autoimmune myocarditis under pembrolizumab treatment for metastatic melanoma
.
J Immunother Cancer
2015
;
3
:
11
.
47.
Alabed
YZ
,
Aghayev
A
,
Sakellis
C
,
Van den Abbeele
AD
. 
Pancreatitis secondary to anti-programmed death Receptor 1 immunotherapy diagnosed by FDG PET/CT
.
Clin Nucl Med
2015
;
40
:
e528
9
.
48.
Manusow
JS
,
Khoja
L
,
Pesin
N
,
Joshua
AM
,
Mandelcorn
ED
. 
Retinal vasculitis and ocular vitreous metastasis following complete response to PD-1 inhibition in a patient with metastatic cutaneous melanoma
.
J Immunother Cancer
2014
;
2
:
41
.
49.
Chan
MM
,
Kefford
RF
,
Carlino
M
,
Clements
A
,
Manolios
N
. 
Arthritis and tenosynovitis associated with the anti-PD1 antibody pembrolizumab in metastatic melanoma
.
J Immunother
2015
;
38
:
37
9
.
50.
Khoja
L
,
Maurice
C
,
Chappell
M
,
MacMillan
L
,
Al-Habeeb
AS
,
Al-Faraidy
N
, et al
Eosinophilic fasciitis and acute encephalopathy toxicity from pembrolizumab treatment of a patient with metastatic melanoma
.
Cancer Immunol Res
2016
;
4
:
175
8
.
51.
Cousin
S
,
Toulmonde
M
,
Kind
M
,
Cazeau
AL
,
Bechade
D
,
Coindre
JM
, et al
Pulmonary sarcoidosis induced by the anti-PD1 monoclonal antibody Pembrolizumab
.
Ann Oncol
2016
;
27
:
1178
9
.
52.
Wilgenhof
S
,
Morlion
V
,
Seghers
AC
,
Du Four
S
,
Vanderlinden
E
,
Hanon
S
, et al
Sarcoidosis in a patient with metastatic melanoma sequentially treated with anti–CTLA-4 monoclonal antibody and selective braf inhibitor
.
Anticancer Res
2012
;
32
:
1355
9
.
53.
Vogel
WV
,
Guislain
A
,
Kvistborg
P
,
Schumacher
TNM
,
Haanen
JBAG
,
Blank
CU
. 
Ipilimumab-induced sarcoidosis in a patient with metastatic melanoma undergoing complete remission
.
J Clin Oncol
2012
;
30
:
e7
10
.
54.
Berthod
G
,
Lazor
R
,
Letovanec
I
,
Romano
E
,
Noirez
L
,
Mazza Stalder
J
, et al
Pulmonary sarcoid-like granulomatosis induced by ipilimumab
.
J Clin Oncol
2012
;
30
:
e156
9
.
55.
Antonia
SJ
,
López-Martin
JA
,
Bendell
J
,
Ott
PA
,
Taylor
M
,
Eder
JP
, et al
Nivolumab alone and nivolumab plus ipilimumab in recurrent small-cell lung cancer (CheckMate 032): a multicentre, open-label, phase 1/2 trial
.
Lancet Oncol
2016
;
17
:
883
95
.
56.
Zibelman
M
,
Ghatalia
P
,
Geynisman
DM
,
Plimack
ER
. 
Checkpoint inhibitors for renal cell carcinoma: current landscape and future directions.
Immunotherapy
2016
;
8
:
785
98
.
57.
Shahabi
V
,
Berman
D
,
Chasalow
SD
,
Wang
L
,
Tsuchihashi
Z
,
Hu
B
, et al
Gene expression profiling of whole blood in ipilimumab-treated patients for identification of potential biomarkers of immune-related gastrointestinal adverse events
.
J Transl Med
2013
;
11
:
75
.
58.
Garon
EB
,
Rizvi
NA
,
Hui
R
,
Leighl
N
,
Balmanoukian
AS
,
Eder
JP
, et al
Pembrolizumab for the treatment of non–small cell lung cancer
.
N Engl J Med
2015
;
372
:
2018
28
.
59.
Attia
P
,
Phan
GQ
,
Maker
AV
,
Robinson
MR
,
Quezado
MM
,
Yang
JC
, et al
Autoimmunity correlates with tumor regression in patients with metastatic melanoma treated with anti-cytotoxic T-lymphocyte antigen-4
.
J Clin Oncol
2005
;
23
:
6043
53
.
60.
Yang
JC
,
Hughes
M
,
Kammula
U
,
Royal
R
,
Sherry
RM
,
Topalian
SL
, et al
Ipilimumab (anti-CTLA4 antibody) causes regression of metastatic renal cell cancer associated with enteritis and hypophysitis
.
J Immunother
2007
;
30
:
825
30
.
61.
Beck
KE
,
Blansfield
JA
,
Tran
KQ
,
Feldman
AL
,
Hughes
MS
,
Royal
RE
, et al
Enterocolitis in patients with cancer after antibody blockade of cytotoxic T-lymphocyte-associated antigen 4, Volume 24
.
J Clin Oncol
2006
;
24
:
2283
9
.
62.
Horvat
TZ
,
Adel
NG
,
Dang
T-O
,
Momtaz
P
,
Postow
MA
,
Callahan
MK
, 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
8
.
63.
Robert
C
,
Schachter
J
,
Long
GV
,
Arance
A
,
Grob
JJ
,
Mortier
L
, et al
Pembrolizumab versus Ipilimumab in advanced melanoma
.
N Engl J Med
2015
;
372
:
2521
32
.
64.
Hua
C
,
Boussemart
L
,
Mateus
C
,
Routier
E
,
Boutros
C
,
Cazenave
H
, et al
Association of Vitiligo with tumor response in patients with metastatic melanoma treated with Pembrolizumab
.
JAMA Dermatol
2016
;
152
:
45
51
.
65.
Sanlorenzo
M
,
Vujic
I
,
Daud
A
,
Algazi
A
,
Gubens
M
,
Luna
SA
, et al
Pembrolizumab cutaneous adverse events and their association with disease progression
.
JAMA Dermatol
2015
;
151
:
1206
12
.
66.
Freeman-Keller
M
,
Kim
Y
,
Cronin
H
,
Richards
A
,
Gibney
G
,
Weber
JS
. 
Nivolumab in resected and Unresectable metastatic melanoma: characteristics of immune-related adverse events and association with outcomes
.
Clin Cancer Res
2016
;
22
:
886
94
.
67.
Johnson
DB
,
Sullivan
RJ
,
Ott
PA
,
Carlino
MS
,
Khushalani
NI
,
Ye
F
, et al
Ipilimumab therapy in patients with advanced melanoma and preexisting autoimmune disorders
.
JAMA Oncol
2016
;
2
:
234
40
.
68.
Lipson
EJ
,
Bodell
MA
,
Kraus
ES
,
Sharfman
WH
. 
Successful administration of ipilimumab to two kidney transplantation patients with metastatic melanoma
.
J Clin Oncol
2014
;
32
:
e69
71
.
69.
Kyi
C
,
Carvajal
RD
,
Wolchok
JD
,
Postow
MA
. 
Ipilimumab in patients with melanoma and autoimmune disease
.
J Immunother Cancer
2014
;
2
:
35
.
70.
Bostwick
AD
,
Salama
AK
,
Hanks
BA
. 
Rapid complete response of metastatic melanoma in a patient undergoing ipilimumab immunotherapy in the setting of active ulcerative colitis
.
J Immunother Cancer
2015
;
3
:
19
.
71.
Davar
D
,
Wilson
M
,
Pruckner
C
,
Kirkwood
JM
. 
PD-1 blockade in advanced melanoma in patients with hepatitis C and/or HIV
.
Case rep Oncol Med
2015
;
2015
:
737389
.
72.
Narita
T
,
Oiso
N
,
Taketomo
Y
,
Okahashi
K
,
Yamauchi
K
,
Sato
M
, et al
Serological aggravation of autoimmune thyroid disease in two cases receiving nivolumab
.
J Dermatol
2016
;
43
:
210
4
.
73.
Lipson
EJ
,
Bagnasco
SM
,
Moore
J
,
Jang
S
,
Patel
MJ
,
Zachary
AA
, et al
Tumor regression and allograft rejection after administration of anti-PD-1
.
N Engl J Med
2016
;
374
:
896
8
.
74.
Downey
SG
,
Klapper
JA
,
Smith
FO
,
Yang
JC
,
Sherry
RM
,
Royal
RE
, et al
Prognostic factors related to clinical response in patients with metastatic melanoma treated by CTL-associated antigen-4 blockade
.
Clin Cancer Res
2007
;
13
:
6681
8
.
75.
Arriola
E
,
Wheater
M
,
Karydis
I
,
Thomas
G
,
Ottensmeier
C
. 
Infliximab for IPILIMUMAB-Related Colitis-Letter
.
Clin Cancer Res
2015
;
21
:
5642
3
.
76.
Arriola
E
,
Wheater
M
,
Krishnan
R
,
Smart
J
,
Foria
V
,
Ottensmeier
C
. 
Immunosuppression for ipilimumab-related toxicity can cause pneumocystis pneumonia but spare antitumor immune control
.
Oncoimmunology
2015
;
4
:
e1040218
.
77.
Spain
L
,
Diem
S
,
Larkin
J
. 
Management of toxicities of immune checkpoint inhibitors
.
Cancer Treat Rev
2016
;
44
:
51
60
.
78.
Naidoo
J
,
Page
DB
,
Li
BT
,
Connell
LC
,
Schindler
K
,
Lacouture
ME
, et al
Toxicities of the anti–PD-1 and anti–PD-L1 immune checkpoint antibodies
.
Ann Oncol
2015
;
26
:
2375
91
.
79.
Patnaik
A
,
Kang
SP
,
Rasco
D
,
Papadopoulos
KP
,
Elassaiss-Schaap
J
,
Beeram
M
, et al
Phase I study of Pembrolizumab (MK-3475; anti–PD-1 monoclonal antibody) in patients with advanced solid tumors
.
Clin Cancer Res
2015
;
21
:
4286
93
.
80.
Maio
M
,
Grob
J-J
,
Aamdal
S
,
Bondarenko
I
,
Robert
C
,
Thomas
L
, et al
Five-year survival rates for treatment-naive patients with advanced melanoma who received ipilimumab plus dacarbazine in a phase III trial
.
J Clin Oncol
2015
;
33
:
1191
6
.
81.
Eggermont
AMM
,
Chiarion-Sileni
V
,
Grob
J-J
,
Dummer
R
,
Wolchok
JD
,
Schmidt
H
, et al
Adjuvant ipilimumab versus placebo after complete resection of high-risk stage III melanoma (EORTC 18071): a randomised, double-blind, phase 3 trial
.
Lancet Oncol
2015
;
16
:
522
30
.