Skip to main content
NIHPA Author Manuscripts logoLink to NIHPA Author Manuscripts
. Author manuscript; available in PMC: 2019 Sep 6.
Published in final edited form as: Curr Rheumatol Rep. 2018 Sep 6;20(10):65. doi: 10.1007/s11926-018-0770-0

Immune-Related Adverse Events in Cancer Patients Treated With Immune Checkpoint Inhibitors

Sabina Sandigursky 1, Adam Mor 2,3,
PMCID: PMC6488223  NIHMSID: NIHMS1022251  PMID: 30191417

Abstract

Purpose of Review

With the advent of cancer immunotherapy and immune checkpoint inhibitors, patients with malignancies can now achieve durable remissions for conditions previously described as terminal. However, immune-related adverse events (irAEs) associated with cancer immunotherapy have become an anticipated consequence of enhanced T cell activation. Through an extensive literature review, we assess the most recent clinical and basic research data concerning immune checkpoint blockade and describe the spectrum of associated irAEs as well as their management.

Recent Findings

Anti-PD-1, anti-PD-L1, and anti-CTLA-4 antibodies are widely used in the management of an array of tumors with incredible clinical remissions. However, irAEs cause significant morbidity and mortality and in some cases, result in withdrawal of cancer therapy and initiation of immunosuppression.

Summary

While this is an exciting time in oncology, irAEs are a barrier to adequate care and therefore deserve close attention and improved capacity to predict and prevent toxicity. Rheumatologists should be familiar with these topics in the eventuality of patient evaluation and management.

Keywords: Cancer, Checkpoint inhibitors, Adverse events, Autoimmune, irAEs

Introduction

The past few years have seen a revolution in cancer treatment. While the concept of cancer immunotherapy is not novel, advances in the field have changed the landscape of cancer treatment. Cancer immunotherapy uses a patient’s own immune system to fight cancer by enhanced activation of the acquired immune response. Immunotherapy approaches can be divided into several categories; adoptive cell transfer (ACT), treatment vaccines, oncolytic viruses, and mono- and bi-specific antibodies including immune checkpoint inhibitors [1, 2]. ACT is a cellular therapy, which involves the removal of immune cells as well as tumor infiltrating lymphocytes from patients, followed by in vitro expansion and subsequent return to the patients where they attack the tumor. Alternatively, patients’ immune cells can be genetically engineered to express tumor-specific receptors, cultured and returned to the patients, which is known as chimeric antigen receptor T cell (CAR-T) therapy. CAR-T therapy is an approved approach to treat B cell malignancies by removing CD19-positive tumor cells from the body. Other cell types that can be used in this way are natural killer (NK) cells, cytotoxic T cells, and dendritic cells (DC). DC therapy provokes anti-tumor responses by causing DC to present tumor antigens to T cells, which activates them to eliminate transformed cells that present the specific antigens. Treatment vaccines boost the immune system’s response to specific antigens expressed on cancer cells and can be used in combination with other anti-cancer approaches for a synergistic effect. Monoclonal antibody technology engineers and generates antibodies against specific tumor antigens, such as those present on tumor cell surfaces in an attempt to detect and eliminate these cells.

Immune checkpoints are regulators of the immune system and are crucial to maintain self-tolerance and to prevent auto-immunity. Some checkpoints, such as CD28, CD40, OX40, and GITR are stimulatory, while others are inhibitory. Inhibitory checkpoints, CTLA-4, LAG3, TIM-3, and PD-1 have been increasingly considered as targets for cancer immunotherapy due to their potential for use in multiple types of tumors [3]. Currently approved immune checkpoint inhibitors (ICIs) block CTLA-4, PD-1, and PD-L1 (a ligand for PD-1). Anti-PD-1 antibodies are designed to bind to PD-1 expressed by tolerant T cells in order to reactivate them and eradicate tumors.

The immunosurveillance paradigm affirms that under homeostatic conditions, the immune system is responsible for clearing malignant cells. It consists of three sequential phases: elimination phase, where malignant cells are removed via the immune system and no overt malignancy is detectable; equilibrium phase, where the quantity of malignant cells and healthy cells are balanced; and escape, where malignant cells overtake healthy tissue and overt cancer is appreciable [4]. Therefore, utilizing the immune system to promote tumor clearance is a logical approach, which has now proven to be an effective cancer treatment. When transformed cells escape immune surveillance and grow into tumor, checkpoint inhibitors are employed to disinhibit the immune system. Not surprisingly, this leads to excessive inflammation, a break in tolerance, and autoimmunity. While this treatment modality has revolutionized cancer treatment, one in every four patients suffers from irAEs, which require immunosuppressive treatment and on occasion, cessation of life saving cancer immunotherapy [5]. This review will discuss checkpoint blockade, indications, irAE pathogenesis, and management.

Regulation of T Cell Activity by Co-receptors

T lymphocytes need engagement of their T cell receptors (TCR) with antigens presented on major histocompatibility complex (MHC) molecules on antigen presenting cells (APC) for activation. This, however, is not sufficient and a second signal is required for complete activation, which can be provided by stimulatory receptors. There are two groups of receptors. The first group is the CD28 superfamily and is composed of CD28 and ICOS, while receptors such as CD27, CD40, CD137 (4–1BB), OX40, and GITR are members of the second group and are part of the TNF receptor superfamily [6]. CD28 is constitutively expressed on approximately 80% of human CD4+ T cells and 50% of CD8+ T cells [7]. Binding to one of its ligands (CD80 or CD86 on APCs), prompts T cell maturation and expansion. An important class of anti-cancer drugs in development engages stimulatory receptors. Agonistic monoclonal antibodies can activate costimulatory signals to enhance T cell priming. Also, they can be used to reinvigorate exhausted T cells in the treatment of cancer as well as infection. For example, the monoclonal CD28 agonist antibody, TGN1412, was evaluated in clinical trials, but severe inflammatory reactions and other toxicities resulted in discontinuation of the trial [8].

Once the T cell is fully activated, it has great potential to inflict significant damage on invading organisms via the immune response. To keep this system from wreaking havoc, signals from inhibitory receptors are required to bring it back to a state of equilibrium. Many of the inhibitory receptors, such as programmed cell death 1 (PD-1), cytotoxic T lymphocyte-associated protein-4 (CTLA-4), as well as others such as, A2AR, B7-H3, BTLA, KIR, LAG3, TIM-3, and VISTA are under investigation as therapeutic targets [3]. Designed to promote tolerance and to prevent autoimmunity, CTLA-4 and PD-1 are critical for terminating the immune response.

In 1987, CTLA-4 was discovered by scientists from the Goldstein laboratory in Marseille (France) looking for cytotoxic cell surface molecules from the cDNA of CD8 T cells [9]. They found that CTLA-4 had high sequence similarity to CD28 and was also located on chromosome 1 [9]. Initially thought to have a role in activation due to high levels of expression on activated cells, it was not until 1995 that James Allison at UC Berkley found that this receptor acts in an inhibitory fashion and directly opposes the function of CD28 [10]. Supporting this notion, it was reported that CTLA-4 knockout mice develop rapidly fatal autoimmunity. Upon T cell activation, CTLA-4 is the first inhibitory molecule to translocate to the cell surface, where it binds to CD80 or CD86. CTLA-4 counteracts the activity of CD28, and binds the same ligands with greater affinity and avidity than CD28, which leads to cell cycle arrest and inhibition of proliferation [1113].

PD-1 was identified in 1992, when Ishida et al. attempted to find the gene responsible for programmed cell death via subtractive hybridization [14]. However, this was not supported experimentally, and the inhibitory function of PD-1 was not revealed until PD-1 null mice developed autoimmunity [15]. PD-1 is a type I transmembrane protein, which exists as a monomer on the cell surface. It has two naturally occurring ligands, programmed death ligand 1 (PD-L1) and programmed death ligand 2 (PD-L2). PD-L1 is found on the surface of many cells, while PD-L2 is restricted to antigen presenting cells (APCs). PD-L2 has a 3-fold higher affinity for PD-1 than PD-L1. Additionally, high levels of PD-1 are present on T cells chronically exposed to abundant antigen, and dysfunctional or exhausted cells are seen in malignancy and chronic infection. Single-nucleotide polymorphisms (SNP) in the pdcd1 gene are associated with autoimmunity (e.g., SLE and MS), cancer (e.g., colon cancer) [16, 17], and chronic infection (e.g., hepatitis B). PD-1 is expressed on immune cells (CD4 and CD8 T cells, NK cells, NKT cells, B cells, macrophages, and DC subsets) upon activation of the immune system and chronic inflammation. The cytoplasmic tail of PD-1 has two tyrosine motifs, an immune tyrosine inhibitory motif (ITIM) and an immune tyrosine stimulating motif (ITSM). The tyrosine phosphatase SHP2 is known to bind to the ITSM of PD-1 and propagate inhibitory signaling downstream [18•].

The activation and inhibition of T cells is a dynamic process, where stimulatory and inhibitory receptors work together to fine-tune the Tcell immune response. The crossroads where all of these molecules are present simultaneously is at the immunological synapse, the interface between the target and the effector T cells [19]. When signals are being transmitted, the APC displaying peptide loaded on major histocompatibility complex (MHC) binds the T cell receptor. If an inhibitory signal is to be propagated, the APC upregulates PD-L1/PD-L2 or CD80/CD86 and binds inhibitory receptors present on effector cells. These discoveries laid the foundation for using immune checkpoint blocking antibodies in the treatment of cancer.

Immune Checkpoint Blocking Antibodies

One of the mechanisms by which tumors evade immunosurveillance is by upregulating inhibitory immune checkpoint ligands [20]. Tumors can use these checkpoint pathways to inhibit immune cells and to protect themselves by inducing tolerance in the invading T cells. Most of the approved checkpoint therapies block inhibitory receptors (Table 1). Blockade of negative feedback signaling to immune cells thus results in an enhanced immune response against tumors.

Table 1.

Monoclonal blocking antibodies

Antibody Drug Indication
CTLA-4 IgG1 Ipilimumab Melanoma
PD-1 IgG4 Nivolumab Melanoma, NSCLC, RCC, HCC, Hodgkin’s lymphoma, SCC of the head and neck, urothelial carcinoma, gastric ca., solid tumors with high MSI, or MRD
PD-1 IgG4 Pembrolizumab Melanoma, NSCLC, primary mediastinal large B cell lymphoma, Hodgkin’s lymphoma, SCC of the head and neck, urothelial carcinoma, gastric ca., solid tumors with high MSI, or MRD
PD-L1 IgG1 Atezolizumab NSCLC, urothelial carcinoma
PD-L1 IgG1 Durvalumab Merkel cell carcinoma, urothelial carcinoma
PD-L1 IgG1 Avelumab Urothelial carcinoma metastatic Merkel cell carcinoma
PD-1 IgG4 and CTLA-4 IgG1 in combination Nivolumab and Ipilimumab Advanced renal cell carcinoma

The first checkpoint antibody approved by the FDA in 2011 was the anti-CTLA-4 (IgG1) antibody ipilimumab for the treatment of melanoma. In a seminal phase III clinical trial, it was demonstrated that ipilimumab was superior for overall survival in metastatic melanoma patients, who had been previously treated with other therapies [21]. Serious irAEs were reported at a rate of 10–15% of patients. In addition, several clinical trials with the anti-CTLA-4 (IgG2) antibody tremelimumab have been completed, but are yet to gain approval and show significance [2224]. Initial clinical trial results with anti-PD-1 (IgG4) antibody nivolumab were published in 2010, while its approval was in 2014 [25]. Nivolumab was initially approved to treat melanoma, lung cancer, kidney cancer, bladder cancer, head and neck cancer, and Hodgkin’s lymphoma. Subsequently, it was approved for additional indications [26]. Pembrolizumab is another anti-PD-1 (IgG4) antibody that was approved to treat melanoma and lung cancer [27]. When compared head to head, pembrolizumab showed clear superiority over ipilimumab in the treatment of advanced melanoma [28•]. Also, adverse events were higher in the ipilimumab group (19.9% vs. 13.3%). In 2016, the anti-PD-L1 (IgG1) antibody atezolizumab was approved for treating bladder cancer [29]. Other anti-PD-L1 antibodies currently in development include avelumab and durvalumab (Table 1). Clinical trials have shown benefits of anti-CTLA-4 therapy also for lung cancer, specifically in combination with other drugs. In ongoing trials, the combination of CTLA-4 blockade with PD-1 or PD-L1 inhibitors is tested on different types of cancer. The combination of anti-PD-1/PD-L1 and anti-CTLA-4 targeting antibodies has been verified in melanoma patients [30]. Treating with concurrent ipilimumab and nivolumab was superior to sequential escalation. However, a higher proportion of patients treated with combination checkpoint blocking antibodies developed serious irAEs. In another study evaluating ipilimumab plus nivolumab or ipilimumab plus placebo, 22% of patients achieved complete response in the combination arm, whereas no complete responses were observed in the placebo group. In patients treated with CTLA-4 blockade, toxicity was observed in up to 60% of patients. Patients receiving anti-PD-1/PD-L1 therapy have more limited toxicity with 24% of patients requiring immunosuppression [31].

Immune checkpoint blockade has improved outcomes for many patients with anti-PD-1 therapy having a better toxicity profile on average, but as with many cancer therapeutics, adverse effects must be managed to allow for greater quality of life and tolerability of treatment.

Immune-Related Adverse Events

The therapeutic success of immune checkpoint blockade comes at a price and many patients develop a wide range of irAEs [5]. Systems that are commonly affected include pulmonary, dermatologic, gastrointestinal, hepatic, and endocrine. Severe and often life-threatening adverse events include pneumonitis and colitis. Overall, drugs targeting the PD-1 pathway cause fewer adverse events than antibodies targeting CTLA-4 [32]. The clinical phenotypes when treated with CTLA-4 versus. PD-1 inhibitors are variable. Patients receiving CTLA-4 inhibitors tend to develop colitis and hypophysitis, while those on PD-1 antagonists can have rarer adverse events such as pneumonitis and thyroiditis [3335].

The mainstay of treatment for irAEs is immunosuppression and interruption of checkpoint inhibitor therapy. Immunosuppressive agents include corticosteroids, mycophenolate mofetil, and tumor necrosis factor-alpha inhibitors (e.g., infliximab). The management approach is based on experience as no prospective clinical trials have addressed this important question. General and organ specific guidelines have been published by a collaborative effort between the American Society of Clinical Oncologists (ASCO) and the National Comprehensive Cancer Network (NCCN) [36••]. irAEs are graded 1 through 4, with 1 being mild, 2 moderate, 3 severe, and 4 life-threatening [36••]. For grade 1, patients are monitored for worsening of the toxicity. For grade 2, checkpoint inhibition should be withheld and not started until the grade drops to 1, and corticosteroids (e.g., prednisone at 0.5 mg/kg/day) should be initiated if the irAEs do not improve within 1 week. For grades 3 and 4, irAEs treatment should be permanently discontinued, and high-dose corticosteroid therapy (prednisone at 1–2 mg/kg/day) initiated until irAEs drop to grade 1 or lower. Interestingly, a very recent study showed that treatment of ipilimumab induced hypophysitis with high-dose corticosteroids is associated with increased mortality in melanoma patients suggesting that management of irAEs is nuanced beyond typical immunosuppression [37•]. Currently, it is unclear whether anti-TNF therapy should be administered before the development of adverse events particularly in the adjuvant setting.

irAEs are thought to be related to the vigorous activation of the immune system, but the exact pathophysiology is unknown. When tolerance is inhibited, autoimmunity ensues. As described, CTLA-4 terminates early activation of immune responses such as T cell priming in lymphoid tissues, while PD-1 is thought to maintain tolerance later and peripherally in the immune response. The differences between these two checkpoints are demonstrated in mouse models [38, 39]. CTLA-4 knockout mice develop overwhelming rapid fatal lymphoproliferative disease. However, PD-1 knockout mice develop arthritis and lupus like features later in life especially when re-derived in C57/B6 background mice [15]. Similarly, patient irAEs profiles are distinct based on whether they receive anti-CTLA-4 or anti-PD-1 therapy. It is unknown whether T cells or autoantibodies mediate the majority of toxicity in irAEs. In some instances, T cell infiltration is evident based on histology (e.g., myocarditis), but in those cases, the TCR were not sequenced to determine whether or not they were to the same tumor-specific antigen. Vitiligo, also commonly seen in melanoma patients undergoing treatment, suggests cross reactivity between the antigen and T cell clones attacking the melanocytes [40].

Other Considerations

After treatment initiation, irAEs may present at any time, but are most frequently seen weeks to months after checkpoint blockade [5]. Since the T cell autoreactive clones are presumably present long after the cessation of treatment, it is possible that autoimmunity may even present many years after treatment. Early (3 weeks onward) toxicity is most commonly dermatologic for both anti-CTLA-4 and anti-PD-1 treatment [33]. It does not appear that the toxicity correlates with medication dosage suggesting that it is not aggregate.

Based on clinical data, only some patients develop irAEs. It is possible that there is genetic predisposition to irAEs as they are known to be important in other autoimmune and rheumatologic diseases. In autoimmunity, genome-wide association studies (GWAS) have been helpful in identifying genes that may predispose to autoimmunity. Perhaps, GWAS studies in cancer patients can identify patients at risk for irAEs. HLA typing may also be of benefit to identify risk factors [41]. Additionally, the microbiome is a hotbed of discussion, when it comes to predicting treatment response as well as risk of colitis with ipilimumab. Two retrospective studies found that patients with high levels of Bacteroidetes phylum have a decreased risk of colitis [42, 43]. Why or how this is happening is unknown, and further studies are warranted to investigate this topic further.

It is intuitive that patients treated with medications that activate the immune system develop off-target effects. However, whether or not this correlates with disease response to treatment is unclear. Two studies have suggested that irAEs that develop in patients treated with ipilimumab or nivolumab correlate with treatment response [31, 33]. While another study by Topalian et al. did not corroborate these findings in patients receiving nivolumab for advanced melanoma [44]. Whether irAEs determine efficacy of ICIs is yet to be determined, but it is unlikely that it is related to tumor type as patients with different types of tumors develop similar toxicity depending on antibody target.

It does not appear that use of immune checkpoint blockade simultaneous with immunosuppression affects the effectiveness of cancer treatment. Retrospective analysis of pooled data from 576 nivolumab treated melanoma patients showed that 71% experienced any grade irAEs and 10% experience grade 3 or 4 irAEs [33]. Of all the patients, 24% received systemic immunosuppression with most irAEs resolving, without affecting the progression-free survival [31].

Data is limited and guidelines recommend cessation of ICIs after a serious adverse event, but perhaps switching between targets may be of value. Retrospective data indicates that patients who developed serious irAEs when treated with ipilimumab can safely be restarted on anti-PD-1 therapy [45]. Three percent of these patients developed irAEs suggesting that toxicity is pathway specific as opposed to all-encompassing for checkpoint inhibitors. While there is significant debate about resuming ICI after developing serious adverse events, it is very clear that ICI are absolutely contraindicated, when they result in life-threatening toxicity such as related to pulmonary, nerve, and cardiovascular systems.

As always, when administering medications, there is a risk versus benefit analysis for every patient. However, it is unclear whether patients with pre-existing autoimmunity would deteriorate by receiving immune checkpoint blockade as those patients were originally excluded from clinical trials. Retrospective studies suggest that these patients can be treated with resultant mild or manageable irAEs [45, 46]. Regarding underlying autoimmune disease, it is possible that some patients may experience transient exacerbations of their disease. Overall, it appears the consensus is that these patients should be considered for treatment with ICI if they have high mortality related malignancy and is advised by the treating physician.

Conclusion

We are now in the midst of a renaissance period of innovation for cancer immunotherapy, which has transformed the field of clinical oncology. Sustained durable responses have provided cures to patients previously diagnosed with a terminal illness. However, response is limited to a subset of patients while a large proportion develops irAEs, which can be severe and life-threatening. Individualized treatment strategies may help to curb overexuberant T cell responses and irAEs. More is to be learned about the underlying mechanisms of irAEs, how to predict who will develop toxicity and how to move forward. Importantly, basic science research efforts should be aimed at understanding the process of this de novo model of human autoimmunity to give insight into other autoimmune-mediated conditions.

Acknowledgments

Funding Information This work was supported by a grant from the NIH (R01 AI25640).

Footnotes

Compliance with Ethical Standards

Conflict of Interest The authors declare that they have no conflict of interest.

Human and Animal Rights and Informed Consent This article does not contain any studies with human or animal subjects performed by any of the authors.

References

Papers of particular interest, published recently, have been highlighted as:

• Of importance

•• Of major importance

  • 1.Weiden J, Tel J, Figdor CG. Synthetic immune niches for cancer immunotherapy. Nat Rev Immunol 2018;18(3):212–9. 10.1038/nri.2017.89. [DOI] [PubMed] [Google Scholar]
  • 2.Farkona S, Diamandis EP, Blasutig IM. Cancer immunotherapy: the beginning of the end of cancer? BMC Med 2016;14:73 10.1186/s12916-016-0623-5. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 3.Pardoll DM. The blockade of immune checkpoints in cancer immunotherapy. Nat Rev Cancer 2012;12(4):252–64. 10.1038/nrc3239. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 4.Dunn GP, Bruce AT, Ikeda H, Old LJ, Schreiber RD. Cancer immunoediting: from immunosurveillance to tumor escape. Nat Immunol 2002;3(11):991–8. 10.1038/ni1102-991. [DOI] [PubMed] [Google Scholar]
  • 5.Postow MA, Sidlow R, Hellmann MD. Immune-related adverse events associated with immune checkpoint blockade. N Engl J Med 2018;378(2):158–68. 10.1056/NEJMra1703481. [DOI] [PubMed] [Google Scholar]
  • 6.Sharma P, Allison JP. Immune checkpoint targeting in cancer therapy: toward combination strategies with curative potential. Cell 2015;161(2):205–14. 10.1016/j.cell.2015.03.030. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 7.Esensten JH, Helou YA, Chopra G, Weiss A, Bluestone JA. CD28 costimulation: from mechanism to therapy. Immunity 2016;44(5): 973–88. 10.1016/j.immuni.2016.04.020. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 8.Suntharalingam G, Perry MR, Ward S, Brett SJ, Castello-Cortes A, Brunner MD, et al. Cytokine storm in a phase 1 trial of the anti- CD28 monoclonal antibody TGN1412. N Engl J Med 2006;355(10):1018–28. 10.1056/NEJMoa063842. [DOI] [PubMed] [Google Scholar]
  • 9.Brunet JF, Denizot F, Luciani MF, Roux-Dosseto M, Suzan M, Mattei MG, et al. A new member of the immunoglobulin superfamily–CTLA-4. Nature 1987;328(6127):267–70. 10.1038/328267a0. [DOI] [PubMed] [Google Scholar]
  • 10.Krummel MF, Allison JP. CD28 and CTLA-4 have opposing effects on the response of T cells to stimulation. J Exp Med 1995;182(2):459–65. 10.1084/jem.182.2.459. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 11.Azuma M, Ito D, Yagita H, Okumura K, Phillips JH, Lanier LL, et al. B70 antigen is a second ligand for CTLA-4 and CD28. Nature 1993;366(6450):76–9. 10.1038/366076a0. [DOI] [PubMed] [Google Scholar]
  • 12.Freeman GJ, Gribben JG, Boussiotis VA, Ng JW, Restivo VA Jr, Lombard LA, et al. Cloning of B7–2: a CTLA-4 counter-receptor that costimulates human T cell proliferation. Science 1993;262(5135):909–11. 10.1126/science.7694363. [DOI] [PubMed] [Google Scholar]
  • 13.Collins AV, Brodie DW, Gilbert RJ, Iaboni A, Manso-Sancho R, Walse B, et al. The interaction properties of costimulatory molecules revisited. Immunity 2002;17(2):201–10. 10.1016/S1074-7613(02)00362-X. [DOI] [PubMed] [Google Scholar]
  • 14.Ishida Y, Agata Y, Shibahara K, Honjo T. Induced expression of PD-1, a novel member of the immunoglobulin gene superfamily, upon programmed cell death. EMBO J 1992;11(11):3887–95. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 15.Nishimura H, Nose M, Hiai H, Minato N, Honjo T. Development of lupus-like autoimmune diseases by disruption of the PD-1 gene encoding an ITIM motif-carrying immunoreceptor. Immunity 1999;11(2):141–51. 10.1016/S1074-7613(00)80089-8. [DOI] [PubMed] [Google Scholar]
  • 16.Tang W, Chen Y, Chen S, Sun B, Gu H, Kang M. Programmed death-1 (PD-1) polymorphism is associated with gastric cardia ad- enocarcinoma. Int J Clin Exp Med 2015;8(5):8086–93. [PMC free article] [PubMed] [Google Scholar]
  • 17.Mojtahedi Z, Mohmedi M, Rahimifar S, Erfani N, Hosseini SV, Ghaderi A. Programmed death-1 gene polymorphism (PD-1.5 C/ T) is associated with colon cancer. Gene 2012;508(2):229–32. 10.1016/j.gene.2012.07.059. [DOI] [PubMed] [Google Scholar]
  • 18.•.Baumeister SH, Freeman GJ, Dranoff G, Sharpe AH. Coinhibitory pathways in immunotherapy for cancer. Annu Rev Immunol 2016;34:539–73. 10.1146/annurev-immunol-032414-112049.Excellent review of coinhibitory pathways targeted for treatment.
  • 19.Davis DM, Dustin ML. What is the importance of the immunological synapse? Trends Immunol 2004;25(6):323–7. 10.1016/j.it.2004.03.007. [DOI] [PubMed] [Google Scholar]
  • 20.Beatty GL, Gladney WL. Immune escape mechanisms as a guide for cancer immunotherapy. Clin Cancer Res 2015;21(4):687–92. 10.1158/1078-0432.CCR-14-1860. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 21.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(8):711–23. 10.1056/NEJMoa1003466. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 22.Sangro B, Gomez-Martin C, de la Mata M, Inarrairaegui M, Garralda E, Barrera P, et al. A clinical trial of CTLA-4 blockade with tremelimumab in patients with hepatocellular carcinoma and chronic hepatitis C. J Hepatol 2013;59(1):81–8. 10.1016/j.jhep.2013.02.022. [DOI] [PubMed] [Google Scholar]
  • 23.Chung KY, Gore I, Fong L, Venook A, Beck SB, Dorazio P, et al. Phase II study of the anti-cytotoxic T-lymphocyte-associated antigen 4 monoclonal antibody, tremelimumab, in patients with refrac- tory metastatic colorectal cancer. J Clin Oncol 2010;28(21):3485–90. 10.1200/JCO.2010.28.3994. [DOI] [PubMed] [Google Scholar]
  • 24.Calabro L, Morra A, Fonsatti E, Cutaia O, Amato G, Giannarelli D, et al. Tremelimumab for patients with chemotherapy-resistant advanced malignant mesothelioma: an open-label, single-arm, phase 2 trial. Lancet Oncol 2013;14(11):1104–11. 10.1016/S1470-2045(13)70381-4. [DOI] [PubMed] [Google Scholar]
  • 25.Brahmer JR, Drake CG, Wollner I, Powderly JD, Picus J, Sharfman WH, et al. Phase I study of single-agent anti-programmed death-1 (MDX-1106) in refractory solid tumors: safety, clinical activity, pharmacodynamics, and immunologic correlates. J Clin Oncol 2010;28(19):3167–75. 10.1200/JCO.2009.26.7609. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 26.Topalian SL, Hodi FS, Brahmer JR, Gettinger SN, Smith DC, McDermott DF, et al. Safety, activity, and immune correlates of anti-PD-1 antibody in cancer. N Engl J Med 2012;366(26):2443–54. 10.1056/NEJMoa1200690. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 27.Goldberg SB, Gettinger SN, Mahajan A, Chiang AC, Herbst RS, Sznol M, et al. Pembrolizumab for patients with melanoma or non- small-cell lung cancer and untreated brain metastases: early analysis of a non-randomised, open-label, phase 2 trial. Lancet Oncol 2016;17(7):976–83. 10.1016/S1470-2045(16)30053-5. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 28.•.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(26):2521–32. 10.1056/NEJMoa1503093.Seminal trial comparing CTLA-4 and PD-1 antagonists in the treatment of advanced melanoma.
  • 29.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(10031):1909–20. 10.1016/S0140-6736(16)00561-4. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 30.Wolchok JD, Kluger H, Callahan MK, Postow MA, Rizvi NA, Lesokhin AM, et al. Nivolumab plus ipilimumab in advanced mel- anoma. N Engl J Med 2013;369(2):122–33. 10.1056/NEJMoa1302369. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 31.Weber JS, Hodi FS, Wolchok JD, Topalian SL, Schadendorf D, Larkin J, et al. Safety profile of nivolumab monotherapy: a pooled analysis of patients with advanced melanoma. J Clin Oncol 2017;35(7):785–92. 10.1200/JCO.2015.66.1389. [DOI] [PubMed] [Google Scholar]
  • 32.Farolfi A, Ridolfi L, Guidoboni M, Nicoletti SV, Piciucchi S, Valmorri L, et al. Ipilimumab in advanced melanoma: reports of long-lasting responses. Melanoma Res 2012;22(3):263–70. [DOI] [PubMed] [Google Scholar]
  • 33.Weber JS, Kahler KC, Hauschild A. Management of immune- related adverse events and kinetics of response with ipilimumab. J Clin Oncol 2012;30(21):2691–7. 10.1200/JCO.2012.41.6750. [DOI] [PubMed] [Google Scholar]
  • 34.Gupta A, De Felice KM, Loftus EV Jr, Khanna S. Systematic re- view: colitis associated with anti-CTLA-4 therapy. Aliment Pharmacol Ther 2015;42(4):406–17. 10.1111/apt.13281. [DOI] [PubMed] [Google Scholar]
  • 35.Ribas A, Puzanov I, Dummer R, Schadendorf D, Hamid O, Robert C, et al. Pembrolizumab versus investigator-choice chemotherapy for ipilimumab-refractory melanoma (KEYNOTE-002): a randomised, controlled, phase 2 trial. Lancet Oncol 2015;16(8): 908–18. 10.1016/S1470-2045(15)00083-2. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 36.••.Brahmer JR, Lacchetti C, Thompson JA. Management of immune- related adverse events in patients treated with immune checkpoint inhibitor therapy: American Society of Clinical Oncology Clinical Practice Guideline Summary. J Oncol Pract 2018;14(4):247–9. 10.1200/JOP.18.00005.American Society of Clinical Oncology treatment guidelines for irAEs.
  • 37.•.Faje AT, Lawrence D, Flaherty K, Freedman C, Fadden R, Rubin K, et al. High-dose glucocorticoids for the treatment of ipilimumab-induced hypophysitis is associated with reduced survival in patients with melanoma. Cancer 2018. 10.1002/cncr.31629. [DOI] [PubMed]
  • 38.Tivol EA, Borriello F, Schweitzer AN, Lynch WP, Bluestone JA, Sharpe AH. Loss of CTLA-4 leads to massive lymphoproliferation and fatal multiorgan tissue destruction, revealing a critical negative regulatory role of CTLA-4. Immunity 1995;3(5):541–7. 10.1016/1074-7613(95)90125-6. [DOI] [PubMed] [Google Scholar]
  • 39.Waterhouse P, Penninger JM, Timms E, Wakeham A, Shahinian A, Lee KP, et al. Lymphoproliferative disorders with early lethality in mice deficient in Ctla-4. Science 1995;270(5238):985–8. [DOI] [PubMed] [Google Scholar]
  • 40.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(1):45–51. 10.1001/jamadermatol.2015.2707. [DOI] [PubMed] [Google Scholar]
  • 41.Chowell D, Morris LGT, Grigg CM, Weber JK, Samstein RM, Makarov V, et al. Patient HLA class I genotype influences cancer response to checkpoint blockade immunotherapy. Science 2018;359(6375):582–7. 10.1126/science.aao4572. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 42.Chaput N, Lepage P, Coutzac C, Soularue E, Le Roux K, Monot C, et al. Baseline gut microbiota predicts clinical response and colitis in metastatic melanoma patients treated with ipilimumab. Ann Oncol 2017;28(6):1368–79. 10.1093/annonc/mdx108. [DOI] [PubMed] [Google Scholar]
  • 43.Dubin K, Callahan MK, Ren B, Khanin R, Viale A, Ling L, et al. Intestinal microbiome analyses identify melanoma patients at risk for checkpoint-blockade-induced colitis. Nat Commun 2016;7: 10391 10.1038/ncomms10391. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 44.Topalian SL, Sznol M, McDermott DF, Kluger HM, Carvajal RD, Sharfman WH, et al. Survival, durable tumor remission, and long- term safety in patients with advanced melanoma receiving nivolumab. J Clin Oncol 2014;32(10):1020–30. 10.1200/JCO.2013.53.0105. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 45.Menzies AM, Johnson DB, Ramanujam S, Atkinson VG, Wong ANM, Park JJ, et al. Anti-PD-1 therapy in patients with advanced melanoma and preexisting autoimmune disorders or major toxicity with ipilimumab. Ann Oncol 2017;28(2):368–76. 10.1093/annonc/mdw443. [DOI] [PubMed] [Google Scholar]
  • 46.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(2): 234–40. 10.1001/jamaoncol.2015.4368. [DOI] [PubMed] [Google Scholar]

RESOURCES