The evolving landscape of immune-related adverse events that follow immune checkpoint immunotherapy in cancer patients

IF 7.5 2区 医学 Q1 IMMUNOLOGY
Alexandra-Chloé Villani
{"title":"The evolving landscape of immune-related adverse events that follow immune checkpoint immunotherapy in cancer patients","authors":"Alexandra-Chloé Villani","doi":"10.1111/imr.13270","DOIUrl":null,"url":null,"abstract":"<p>Immune checkpoint inhibitors (ICIs), including antibodies targeting anti-cytotoxic T lymphocyte antigen 4 (CTLA-4), anti-programmed cell death 1 (PD-1), anti-programmed cell death 1 ligand 1 (PD-L1) have revolutionized cancer treatment.<span><sup>1, 2</sup></span> Both PD-1 and CTLA-4 are upregulated upon T-cell activation and function to inhibit peripheral T-cell responses. These receptors maintain immune homeostasis, limiting tissue damage during infection and helping to prevent autoimmune disease. CTLA-4 suppresses T-cell activation by cell-extrinsic competition with CD28 for binding the same ligands and cell-intrinsic signaling mechanisms.<span><sup>3</sup></span> The PD-1:PD-:L1/L2 pathway also suppresses T-cell responses by inhibiting TCR/CD28 signaling, target cell lysis and cytokine production.<span><sup>4</sup></span> PD-1 ligand is expressed across a diverse set of hematopoietic and non-hematopoietic cells, suggesting it may have more impact in peripheral tissues, whereas CTLA-4 and its ligands are primarily expressed in secondary lymphoid organs.<span><sup>3</sup></span> Collectively, ICI immunotherapy leverages this latter biology to regulate T-cell activation and tolerance, allowing the immune system to overcome cancer's evasion of immunity and reestablish its ability to attack tumor tissues.<span><sup>1, 2</sup></span> CTLA-4 was the first target to be approved for ICI therapy (i.e., Ipilimumab) in 2011<span><sup>5-8</sup></span> to treat advanced melanoma. Since then, eight agents targeting PD-1/PD-L1 or CTLA-4 pathways have been approved by the Food and Drug Administration (FDA) for over 20 cancer types across 80 indications.<span><sup>9</sup></span> Importantly, the utilization of ICIs in the adjuvant setting is also on the rise, resulting in further growth of the number patients receiving ICI therapy. Cancer patients treated with ICIs are now anticipated to live longer; but prolonged exposure to ICI treatment also puts them at greater risk of long-term complications associated with this line of therapy.<span><sup>10-14</sup></span></p><p>Unfortunately, the success of ICIs is limited by the emergence of treatment-induced inflammatory toxicities, termed immune-related adverse events (irAEs). irAEs can affect any organ system in the body with a range of severity, resulting in significant morbidity for patients and considerable cost to the healthcare system. These irAEs can develop during or after treatment and often require immunosuppressive therapy or cessation of ICI treatment, thereby limiting the potential lifesaving benefits from ICI therapy.<span><sup>1, 2, 15, 16</sup></span> Depending on the treatment regimen (i.e., drug types, combination, and doses), 66%–96% of patients develop irAEs, and 7%–59% experience severe irAEs.<span><sup>17, 18</sup></span> Fulminant and fatal irAEs—the most common being myocarditis/myositis, pneumonitis, and neurologic events (e.g., Guillain–Barre syndrome and myasthenia gravis)—are experienced by approximately 0.4% of cancer patients treated with anti-PD-1/PD-L1 monotherapy, and 1.2% of patients treated with combination PD-1/CTLA-4 therapy.<span><sup>19, 20</sup></span> Toxicities can manifest at any point during the treatment, but they tend to occur most frequently within the initial 3 months following the initiation of ICI.<span><sup>15, 16</sup></span> Immuno-oncology remains a rapidly evolving field, with many more co-inhibitory pathways beyond CTLA-4 and PD-1 likely to become valuable therapeutic targets in the near future. For example, relatlimab, a compound that blocks lymphocyte antigen 3 (LAG-3) has recently been approved for use in combination with anti-PD1 therapy.<span><sup>21</sup></span> This target is predominantly found in exhausted T cells and could potentially address T cell-related anergy associated with the tumor microenvironment. As ICIs become first- and second-line cancer treatment for more tumor types in the years to come, together with the increased use of ICIs in the adjuvant setting as well as in combination therapy to increase the anti-tumor response efficacy and overcome treatment resistance,<span><sup>22, 23</sup></span> it is expected that the incidence of irAEs will continue to rise and severely limit the life-saving potential of ICIs.</p><p>Intriguingly, irAEs occurring in the setting of ICI therapy closely resemble idiopathic autoimmune diseases, including rheumatoid arthritis (RA), autoimmune hepatitis, and inflammatory bowel disease.<span><sup>1</sup></span> Further, polymorphisms of PD-1 and CTLA-4 are associated with various autoimmune disease conditions—such as Type 1 diabetes, thyroiditis, Graves' disease, celiac disease, systemic lupus erythematosus, and rheumatoid arthritis<span><sup>15, 16, 24, 25</sup></span>—with which the observed treatment-related irAEs can share clinical features. irAEs are treated with many of the same therapeutic interventions as autoimmune diseases, including steroids, biologics agents, and sometimes surgery. Insights into the pathogenesis of irAEs, a setting in which the exact disease trigger and timing of disease initiation are known, may thus have implications not only for the care of cancer patients but also for our understanding of idiopathic autoimmune diseases. Studying the cellular and associated molecular patterns associated with irAE may provide a rare window into the exact moment when a patient's own immune system starts mounting an inflammatory response against normal tissue. Such study may also enable generating novel insights into the mechanisms of immune regulation throughout the body and could further our understanding of how CTLA-4 and PD-1 function to maintain immune tolerance.<span><sup>26</sup></span> As such, deciphering irAE pathogenesis may have implications not only for the care of cancer patients but also provide critical insights into the earliest steps driving and sustaining spontaneous autoimmune diseases, which are challenging to discern and study when such diseases are fully established.</p><p>While we can hypothesize that the trigger of the irAE onset is the manipulation of T-cell signaling pathways and the breaking of tissue-specific immune tolerance,<span><sup>1, 2, 15, 16</sup></span> the downstream mechanisms involved in driving and sustaining excessive immune activation against normal tissue remain poorly understood. This is in part due to the fact that preclinical animal models have yet to recapitulate the full spectrum of irAE clinical presentations.<span><sup>15, 16</sup></span> Examples of disease mechanisms proposed thus far have included shared antigens between tumor and affected tissue, preexisting subclinical inflammation, preexisting autoantibodies, antibody-dependent cytotoxicity, and environmental insults.<span><sup>15, 16, 26, 27</sup></span> Given the heterogeneity and breadth of irAE clinical presentations, it is likely that irAEs affecting different organ systems have different pathological drivers. Among the irAEs sharing similar histological characteristics within a specific tissue, it is also possible that multiple mechanistic endotypes result in comparable clinical manifestations. Our limited mechanistic understanding of irAEs to ICI has severely restricted the development of targeted treatment solutions. Hitherto, treatment management guidelines for irAE have been developed almost entirely based on small retrospective clinical studies and expert opinion. Because the mechanisms driving toxicity and severity across different organ systems will likely differ, it will require that the medical community conceive prospective treatment trials for each irAE individually, as opposed to studying multiple irAEs together. Over the last few years, limited progress has been made in understanding the biological drivers of ICI-related irAEs, and in developing effective mitigation strategies. Further research into the cellular and molecular underpinning of irAEs will be critical to define better predictors of toxicity to enable the prevention of their occurrence, as well as the development of new, targeted, and improved treatment solutions that can mitigate these toxicities while preserving the anti-tumor immune response.</p><p>This issue comprises a compilation 13 review pieces from top scientists and medical experts in the field who discuss our current understanding of these ICI associated inflammatory toxicities across organ systems. Reviews include several in-depth overviews of tissue-specific irAE presentations. They address our current understanding of cellular and molecular drivers of irAEs and their relationship to anti-tumor immune response, including discussions on available preclinical models to study these mechanisms. They cover potential predictive biomarkers and genetic risk factors identified to-date, and discuss current management strategies being used, as well as clinical trials being developed using therapeutic candidates intended to limit irAE onset and improve patient outcomes. Collectively, these reviews summarize the state-of-the field of this evolving and complex subject, the critical challenges to ICI therapies posed by irAEs, along with the many opened questions and the strategies that should be the focus of future investigations.</p><p>The first set of six reviews delves deeply into our understanding of organ-specific irAEs. It is crucial to better understand the immunopathology of irAEs across organ-systems to allow for a more personalized immune suppression therapy and to spare negative effects on anti-tumor immunity. Although irAEs can target any organ system, toxicities occur more frequently at barrier tissues, including the gastrointestinal tract, skin, liver, and the lungs,<span><sup>15, 16, 26</sup></span> and these are the focus of the first three reviews. The fact that barrier organs are so commonly involved suggests that the antigenic targets of the immune response may be the commensal microbiome, though this has not yet been demonstrated.</p><p>One of the first irAEs reported was vitiligo, which is a condition characterized by immune-mediated destruction of melanocytes that can occur in cancer patients with melanoma undergoing going treatment with various immunotherapies.<span><sup>15, 16, 26</sup></span> Vitiligo is driven by T cells and antibodies targeting melanocyte antigens, which are also produced by melanoma. This immune response leads to the destruction of melanocytes, resulting in the characteristic depigmentation observed in affected individuals.<span><sup>28</sup></span> Given these early observations, it was originally hypothesized that other irAEs might also result from targeting antigens common to healthy and malignant tissue, similarly to the mechanisms observed in vitiligo. Nonetheless, the fact that the majority of irAEs are not specific to a particular tumor type cast doubt on this hypothesis. Alternative hypotheses have now emerged that are discussed in the first three reviews. These posit a role for tissue-resident memory cells (TRM cells), which are long-lived memory effectors that reside in peripheral tissues and make up the majority of T cells in barrier tissue. TRM cells express PD-1 and other immune checkpoint molecules that control their activation. These cells play important roles in tissue surveillance and recall responses to pathogens.<span><sup>29-31</sup></span> Recent studies have investigated whether ICI therapy triggers irAE by promoting the expansion and activation of local resident TRM cells. This work is discussed in the first three reviews covering irAE affecting the gastrointestinal track, the skin, and the lung.</p><p>The review by Michael Dougan provides a comprehensive review on the gastrointestinal (GI) toxicities caused by ICIs.<span><sup>32</sup></span> Although most of the GI irAEs are mild in nature, their high occurrence rate makes them the leading cause of severe toxicities associated with ICIs. In his review, he provides an in-depth description of the clinical manifestations along with a thorough discussion of the immune mechanisms known to-date (e.g., gut CD8<sup>+</sup> TRMs) associated with ICI-colitis, which is the most common GI toxicity associated with the use of ICIs.<span><sup>33</sup></span> He also discusses risk factors associated with ICI-colitis, current standard of care, upcoming clinical trials, and the challenges related to the current lack of clinical trial data to guide management of patient care. Importantly, he highlights multiple unanswered questions in the field that require further investigation.</p><p>The second review from Blair Allais and colleagues is focused on dermatologic toxicities, which are the most frequently reported, the most visible, and among the earliest seen in cancer patients on ICI therapy.<span><sup>34</sup></span> While most of these cutaneous immune-related adverse effects (cirAEs) are mild, severe presentations occur.<span><sup>35</sup></span> Allais et al. provide a clear and thorough discussion of what is known about cirAEs through the different lenses being used to study them. These include harnessing the power of large datasets to understand clinical presentations and their association with outcomes. They establish specific definitions for each cirAEs, and their need for more targeted therapies. They describe leveraging cellular and molecular strategies to infer novel insights into disease mechanisms. They also describe the current understanding of immunosuppression treatment for managing cirAEs, as well as future opportunities in the field to help improve patient care.</p><p>Fiamma Berner and Lukas Flatz focused their review on two emerging classes of self-antigens involved in skin toxicity and pneumonitis in non-small cell lung cancer patients undergoing ICI therapy.<span><sup>36</sup></span> They discuss how these self-antigen-specific T cells are believed to play a dual role by mediating irAE to skin and lung, and by promoting anti-tumor immune responses. Such dual association creates a challenge in distinguishing between the immune signature associated with irAE from those associated with treatment response. The review also highlights methods for the identification of self-antigen targets and discusses the role of antigens shared by organs affected by irAEs and tumors. Finally they discuss current challenges and solutions that may lead to successful cancer patient treatment with ICIs in the absence of problematic irAEs.</p><p>Gary Reynolds delves into rheumatological irAEs, which are disabling, can limit treatment efficacy and evolve to become chronic.<span><sup>37</sup></span> These presentations can superficially share features of common immune-mediated rheumatic diseases (IMRDs),<span><sup>38</sup></span> such as rheumatoid arthritis, psoriatic arthritis, Sjogren's syndrome, and polymyalgia rheumatica. This review discusses the parallels and differences between rheumatic irAEs and IMRDs, and highlight that, based on current knowledge, they are clinically and immunologically distinct entities.</p><p>Harish Seethapathy, Kavita Mistry, and Meghan Sise elegantly describe irAE manifestations in the kidney, which present as acute interstitial nephritis (ICI-AIN).<span><sup>39</sup></span> They thoroughly explore the clinical manifestation of ICI-AIN and highlight its distinctive presentation compared to conventional drug-induced AIN. The authors provide a detailed description of current mechanistic understanding of ICI-AIN, and underscore the diagnostic and therapeutic implications that arise from these findings.</p><p>To conclude this first section of the issue, Jingyi Gong, Tomas Neilan, and Daniel A. Zlotoff present a thorough review on immune-related myocarditis (irMyocarditis), which, among all the cardiovascular toxicities, has been the most extensively studied due to its substantial risk of mortality.<span><sup>40</sup></span> While irMyocarditis occurs in 1% of ICI recipients, the incidence is expected to continue rising with the increasing use of combination regimens to treat cancer patients.<span><sup>19-21, 41-43</sup></span> It is fatal in 20%–40% of cases, which is approximately 10-fold higher than in non-ICI lymphocytic myocarditis.<span><sup>41-44</sup></span> Nearly half of the cases result in major adverse cardiac events, such as severe arrhythmias, cardiogenic shock, and sudden cardiac death.<span><sup>41-44</sup></span> Furthermore, the development of irMyocarditis is a near-absolute contraindication for future use of ICI therapy, which in turn may harm the oncologic prognosis.<span><sup>45</sup></span> The authors frame their review around two primary objectives. First, they provide a comprehensive summary of the current understanding of irMyocarditis pathogenesis, and they compare it to other types of myocardial inflammation, which they examine across three categories: the cellular mediators, participating molecular signals and soluble factors, and T-cell receptor (TCR) clonality and specificity. Second, they discuss different hypotheses explaining its heightened morbidity compared to other forms of cardiac inflammation and why it only affects a limited number of cancer patients receiving ICI therapy. They conclude their elegant review by highlighting the many knowledge research and clinical gaps in the field requiring further investigation to enable developing better solutions for patient care.</p><p>While the above six reviews each focus on the clinical manifestations associated with specific organ system irAEs, the next section includes four reviews that delve more deeply into our biological understanding of irAE presentations across tissues, the known cellular drivers, their relationship to autoimmunity, and the available preclinical models that can be leveraged to study their mechanistic underpinnings. Importantly, defining the underlying disease mechanisms presents an opportunity not only to alleviate these detrimental side effects but to also improve the efficacy of ICI therapy itself.</p><p>Namrata Singh, Anne Hocking, and Jane Buckner provide an interesting review on the intersection between irAEs to ICI and autoimmune diseases.<span><sup>46</sup></span> While several irAEs can phenocopy the clinical presentation of well-defined autoimmune diseases, it remains unclear whether these diseases are truly the same syndrome, especially as in many cases the ICI-associated presentation is more fulminate.<span><sup>47</sup></span> This review explores whether the mechanisms responsible for inducing ICI-related irAEs are similar or different from those underlying spontaneous autoimmunity, focusing on ICI-diabetes, ICI-thyroiditis, and ICI-arthritis. The authors present a summary of our current understanding of the role of genetics and autoantibodies in the development of irAEs, and discusses the use of single-cell genomic strategies that have enabled the identification of specific T-cell signatures associated with irAEs. Ultimately, deciphering the relationship between these irAEs and spontaneous autoimmune diseases holds significant importance in terms of understanding basic mechanisms and clinical implications. Indeed, from a basic science standpoint, elucidating the connection between these clinical presentations will generate valuable insights into the fundamental mechanisms of autoimmunity and the biological principles of immune tolerance in human. From a clinical perspective, unraveling the relationship between irAEs and autoimmunity could help nominate therapeutic strategies for irAE based on therapies already clinically approved to mitigate autoimmunity.</p><p>Kavita Dhodapkar, Alyssa Duffy, and Madhav Dhodapkar discuss how the use of immune checkpoint inhibitor therapy can impact B-cell tolerance and drive alterations in the B-cell compartments.<span><sup>48</sup></span> Their review explores the potential involvement of humoral immunity, and the role of specific B-cell subsets and autoantibodies in driving the irAE pathogenesis associated with the use of ICI. They also highlight the need to invest more resources in investigating the cross talk between T and B cells that drives the activation of pathogenic B cells, contributing to the development of irAEs.</p><p>Noah Earland and colleagues lay out a thorough perspective<span><sup>49</sup></span> on their recently published work that analyzed a cohort of advanced melanoma using a multimodal strategy involving the use of single-cell RNA sequencing, as well as mass cytometry of peripheral blood cells to identify features predictive of irAE development.<span><sup>50</sup></span> Their work showed that increased TCR diversity together with increased circulating levels of activated CD4 effector memory T cells are both associated with the development of severe irAEs, independent of the tissue affected.<span><sup>50</sup></span> Through their review, they contextualize their results in comparison to other published work on irAE blood-based biomarkers. They examine possible mechanistic links between CD4 memory T cells and effector memory T cells, along with other factors that could contribute to irAE development, such as autoreactive tissue-resident memory T cells located at the tissue site of toxicity. Finally, they discuss factors that should be considered to empower future clinical translation of findings to develop and establish blood-based irAE-related clinical tests.</p><p>Presently, the pathophysiological mechanisms of irAE are poorly understood in part because preclinical mouse models underestimate the occurrence of irAEs compared to patient experience,<span><sup>51</sup></span> or do not accurately replicate critical aspects of the human irAE biology.<span><sup>15, 16, 26, 52-54</sup></span> Nevertheless, there is a multitude of available animal systems that could be tailored and leveraged to further model biological pathways identified in patients. For the last manuscript of this section, Morgan Cina, Jessica Venegas, and Arabella Young put together a very comprehensive review on the preclinical models available that capture some aspects of the irAE etiology across different organ systems. These can be used to study the biological underpinnings of irAEs, along with their relationship to anti-tumor immune responses.<span><sup>55</sup></span> They examine the contexts for which these models are most useful, how they could be improved, as well as the advantages and disadvantages of different model systems. Approaches include in silico modeling of published datasets, in vitro organoid models, in vivo mouse and non-human primate models, as well as canine patients. They review why the development of model systems to study irAEs has not progressed rapidly. Particularly in comparison to efforts invested in establishing models to study therapeutic combination strategies. The authors end by discussing what the future holds for developing new preclinical models that capture with higher fidelity the breadth irAE presentations. Better models are critical to ultimately improve our mechanistic understanding of irAEs and their associated treatments.</p><p>The third and last section of this issue includes three reviews that examine what factors have been identified to-date that may help determine who will develop irAEs to ICI, whether we can predict which patient is predisposed to irAEs, and what is the relationship between irAE presentations and anti-tumor immune response. The ability to predict the occurrence of specific irAEs to ICI will be a critical step towards the development of prophylactic therapies and could prove valuable in making informed decisions regarding tailoring cancer treatment options for each patient.</p><p>One of the strategies used to identify disease predictors is to focus on uncovering germline susceptibility variants to ICI-related irAEs, which is elegantly reviewed in this issue by Alexander Gusev.<span><sup>56</sup></span> Because these irAEs can phenocopy the clinical presentation of some sporadic autoimmune diseases, it has been hypothesized that they could be linked to autoimmunity through germline genetics and would be heritable.<span><sup>57-59</sup></span> In this review, he summarizes current work mapping susceptibility germline variants to irAEs, outcome, and responses to ICIs. He also provides perspectives on recently published work from his group that leverages genome-wide association analysis to identify IL7 susceptibility variants associated with irAEs,<span><sup>60</sup></span> findings that were subsequently independently replicated.<span><sup>61</sup></span> He also discusses extant questions in the field, as well as opportunities to leverage germline genetic association studies to further our understanding of irAE etiology, in addition to improving our ability to predicting them.</p><p>There are currently no FDA approved biomarkers that predict the occurrence of irAEs, making it challenging to identify cancer patients for whom the risk of suffering from toxicity to ICI therapy is outweighed by the potential lifesaving benefits of receiving them. Moreover, the timing of these toxicities is not as predictable as side effects associated with other cancer treatments, such as chemotherapy and targeted therapy.<span><sup>62</sup></span> Nevertheless, several efforts to map such biomarkers for irAE toxicities that could guide treatment selection, prevention, and monitoring have been initiated. These are thoroughly review by Rachel Goodman, Seungyeon Jung, Justin Balko, and Douglas Johnson.<span><sup>63</sup></span> The authors describe different molecular and clinical biomarkers across three categories, including those that can help: (i) risk stratify patient pre-treatment to identify subjects likely to develop toxicities; (ii) diagnose irAEs, which remain challenging to do in many cases; and (iii) predict occurrence of irAEs during treatment course. These could enable tailoring clinical decision-making, including whether to alter treatment course or to increase the frequency of patient monitoring.</p><p>We conclude this issue with a comprehensive review by Steven M. Blum, Sherin Rouhani, and Ryan Sullivan on the molecular and clinical relationship between anti-tumor immune response and irAEs, along with a discussion of the role that irAE treatments may play in modulating the anti-tumor immune response.<span><sup>64</sup></span> They examine the relationship between irAEs and anti-tumor immunity, which can be complex<span><sup>65</sup></span> and influenced by factors such as the specific irAE-affected organ, tumor histology, and individual patient's characteristics. The authors highlight the importance of developing therapeutic strategies that can maximize the anti-tumor immunity efficacy of ICIs while minimizing the occurrence of irAEs by modulating both the host and the tumor immune environments, which will ultimately be essential to improve patient outcomes.</p><p>The landscape of cancer patient populations being treated with ICI therapies has significantly changed since their FDA approval for the treatment of metastatic melanoma in 2011; ICI is now used in indications with curative intent. For malignancies like metastatic melanoma, which were largely fatal before the introduction of ICI therapy, the benefits of ICIs far surpass the potential risks of developing irAEs in most patients, making such risk acceptable. In the context of adjuvant and neoadjuvant therapy, the risk-to-benefit ratio is less straightforward; while there has been improved disease-free survival, it is important to note that a considerable proportion of patients would be expected to achieve cancer remission without the use of ICIs, highlighting the need to reevaluate the irAEs risk versus the reward of being cancer-free. The illustration included in this introduction (Figure 1) captures the complicated balance between the benefits and risks associated with the use of ICI therapy to treat cancer patient. Despite the emergence of irAEs, it is important to remember that the introduction of ICI drugs has resulted in many good news stories, providing new hopes to cancer patients.</p><p>With the increasing utilization of ICI therapy among the cancer patient population, it is becoming increasingly critical to support research efforts aimed at understanding the root cause of irAE development, and how they can be mitigated while promoting anti-tumor immunity. Reviews included in this issue highlight how our understanding of irAEs has evolved over the years, along with the many challenges to be overcome and the remaining opened questions to be answered in the field, such as, which factors play a role in determining patient risk of experiencing irAEs? Can we predict which patient will develop irAEs? What factors contribute to the higher incidence of irAEs in certain organ systems? How are spontaneous autoimmune diseases and irAEs that phenocopy them related? Which factors influence the severity of an irAE in a particular patient, which can range from transient to chronic or fatal? How can we choose the most effective treatment strategies to treat irAEs while sparing the anti-tumor immune response? Once an irAE has improved upon treatment, which patient is at risk for redeveloping irAEs and which patient can be safely rechallenged with ICI therapy? Answering these outstanding questions will likely be the focus of upcoming studies in the immuno-oncology field.</p><p>Lastly, a broad theme that emerges from several reviews in this issue is the need to provide more financial support to this research across disciplines and to cross-institutional multidisciplinary teams spanning healthcare systems, academic researchers, federal research agencies, commercial partners, and patient advocacy groups to solve the underpinnings of irAEs and integrate critical discoveries into sustainable clinical solutions and applications. Even once irAE therapeutic targets are identified, the field will still necessitate cross-institutional collaborative endeavors to establish the clinical trial infrastructure required for evaluating these treatments. We owe it to cancer patients to work together as a community to find solutions to mitigate irAEs in order to improve their quality of life and enable them to optimally benefit from the life-saving virtues of ICI-therapy to fight their cancers.</p><p>ACV has been a paid consultant to Bristol Myers Squibb.</p><p>ACV is funded by the National Institute of Health Director's New Innovator Award (DP2CA247831), the Damon Runyon-Rachleff Innovation Award, The Melanoma Research Alliance Young Investigator Award (https://doi.org/10.48050/pc.gr.143739), the Massachusetts General Hospital (MGH) Transformative Scholar in Medicine Award, and the MGH Howard M. Goodman Fellowship.</p>","PeriodicalId":178,"journal":{"name":"Immunological Reviews","volume":"318 1","pages":"4-10"},"PeriodicalIF":7.5000,"publicationDate":"2023-08-26","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://onlinelibrary.wiley.com/doi/epdf/10.1111/imr.13270","citationCount":"0","resultStr":null,"platform":"Semanticscholar","paperid":null,"PeriodicalName":"Immunological Reviews","FirstCategoryId":"3","ListUrlMain":"https://onlinelibrary.wiley.com/doi/10.1111/imr.13270","RegionNum":2,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":null,"EPubDate":"","PubModel":"","JCR":"Q1","JCRName":"IMMUNOLOGY","Score":null,"Total":0}
引用次数: 0

Abstract

Immune checkpoint inhibitors (ICIs), including antibodies targeting anti-cytotoxic T lymphocyte antigen 4 (CTLA-4), anti-programmed cell death 1 (PD-1), anti-programmed cell death 1 ligand 1 (PD-L1) have revolutionized cancer treatment.1, 2 Both PD-1 and CTLA-4 are upregulated upon T-cell activation and function to inhibit peripheral T-cell responses. These receptors maintain immune homeostasis, limiting tissue damage during infection and helping to prevent autoimmune disease. CTLA-4 suppresses T-cell activation by cell-extrinsic competition with CD28 for binding the same ligands and cell-intrinsic signaling mechanisms.3 The PD-1:PD-:L1/L2 pathway also suppresses T-cell responses by inhibiting TCR/CD28 signaling, target cell lysis and cytokine production.4 PD-1 ligand is expressed across a diverse set of hematopoietic and non-hematopoietic cells, suggesting it may have more impact in peripheral tissues, whereas CTLA-4 and its ligands are primarily expressed in secondary lymphoid organs.3 Collectively, ICI immunotherapy leverages this latter biology to regulate T-cell activation and tolerance, allowing the immune system to overcome cancer's evasion of immunity and reestablish its ability to attack tumor tissues.1, 2 CTLA-4 was the first target to be approved for ICI therapy (i.e., Ipilimumab) in 20115-8 to treat advanced melanoma. Since then, eight agents targeting PD-1/PD-L1 or CTLA-4 pathways have been approved by the Food and Drug Administration (FDA) for over 20 cancer types across 80 indications.9 Importantly, the utilization of ICIs in the adjuvant setting is also on the rise, resulting in further growth of the number patients receiving ICI therapy. Cancer patients treated with ICIs are now anticipated to live longer; but prolonged exposure to ICI treatment also puts them at greater risk of long-term complications associated with this line of therapy.10-14

Unfortunately, the success of ICIs is limited by the emergence of treatment-induced inflammatory toxicities, termed immune-related adverse events (irAEs). irAEs can affect any organ system in the body with a range of severity, resulting in significant morbidity for patients and considerable cost to the healthcare system. These irAEs can develop during or after treatment and often require immunosuppressive therapy or cessation of ICI treatment, thereby limiting the potential lifesaving benefits from ICI therapy.1, 2, 15, 16 Depending on the treatment regimen (i.e., drug types, combination, and doses), 66%–96% of patients develop irAEs, and 7%–59% experience severe irAEs.17, 18 Fulminant and fatal irAEs—the most common being myocarditis/myositis, pneumonitis, and neurologic events (e.g., Guillain–Barre syndrome and myasthenia gravis)—are experienced by approximately 0.4% of cancer patients treated with anti-PD-1/PD-L1 monotherapy, and 1.2% of patients treated with combination PD-1/CTLA-4 therapy.19, 20 Toxicities can manifest at any point during the treatment, but they tend to occur most frequently within the initial 3 months following the initiation of ICI.15, 16 Immuno-oncology remains a rapidly evolving field, with many more co-inhibitory pathways beyond CTLA-4 and PD-1 likely to become valuable therapeutic targets in the near future. For example, relatlimab, a compound that blocks lymphocyte antigen 3 (LAG-3) has recently been approved for use in combination with anti-PD1 therapy.21 This target is predominantly found in exhausted T cells and could potentially address T cell-related anergy associated with the tumor microenvironment. As ICIs become first- and second-line cancer treatment for more tumor types in the years to come, together with the increased use of ICIs in the adjuvant setting as well as in combination therapy to increase the anti-tumor response efficacy and overcome treatment resistance,22, 23 it is expected that the incidence of irAEs will continue to rise and severely limit the life-saving potential of ICIs.

Intriguingly, irAEs occurring in the setting of ICI therapy closely resemble idiopathic autoimmune diseases, including rheumatoid arthritis (RA), autoimmune hepatitis, and inflammatory bowel disease.1 Further, polymorphisms of PD-1 and CTLA-4 are associated with various autoimmune disease conditions—such as Type 1 diabetes, thyroiditis, Graves' disease, celiac disease, systemic lupus erythematosus, and rheumatoid arthritis15, 16, 24, 25—with which the observed treatment-related irAEs can share clinical features. irAEs are treated with many of the same therapeutic interventions as autoimmune diseases, including steroids, biologics agents, and sometimes surgery. Insights into the pathogenesis of irAEs, a setting in which the exact disease trigger and timing of disease initiation are known, may thus have implications not only for the care of cancer patients but also for our understanding of idiopathic autoimmune diseases. Studying the cellular and associated molecular patterns associated with irAE may provide a rare window into the exact moment when a patient's own immune system starts mounting an inflammatory response against normal tissue. Such study may also enable generating novel insights into the mechanisms of immune regulation throughout the body and could further our understanding of how CTLA-4 and PD-1 function to maintain immune tolerance.26 As such, deciphering irAE pathogenesis may have implications not only for the care of cancer patients but also provide critical insights into the earliest steps driving and sustaining spontaneous autoimmune diseases, which are challenging to discern and study when such diseases are fully established.

While we can hypothesize that the trigger of the irAE onset is the manipulation of T-cell signaling pathways and the breaking of tissue-specific immune tolerance,1, 2, 15, 16 the downstream mechanisms involved in driving and sustaining excessive immune activation against normal tissue remain poorly understood. This is in part due to the fact that preclinical animal models have yet to recapitulate the full spectrum of irAE clinical presentations.15, 16 Examples of disease mechanisms proposed thus far have included shared antigens between tumor and affected tissue, preexisting subclinical inflammation, preexisting autoantibodies, antibody-dependent cytotoxicity, and environmental insults.15, 16, 26, 27 Given the heterogeneity and breadth of irAE clinical presentations, it is likely that irAEs affecting different organ systems have different pathological drivers. Among the irAEs sharing similar histological characteristics within a specific tissue, it is also possible that multiple mechanistic endotypes result in comparable clinical manifestations. Our limited mechanistic understanding of irAEs to ICI has severely restricted the development of targeted treatment solutions. Hitherto, treatment management guidelines for irAE have been developed almost entirely based on small retrospective clinical studies and expert opinion. Because the mechanisms driving toxicity and severity across different organ systems will likely differ, it will require that the medical community conceive prospective treatment trials for each irAE individually, as opposed to studying multiple irAEs together. Over the last few years, limited progress has been made in understanding the biological drivers of ICI-related irAEs, and in developing effective mitigation strategies. Further research into the cellular and molecular underpinning of irAEs will be critical to define better predictors of toxicity to enable the prevention of their occurrence, as well as the development of new, targeted, and improved treatment solutions that can mitigate these toxicities while preserving the anti-tumor immune response.

This issue comprises a compilation 13 review pieces from top scientists and medical experts in the field who discuss our current understanding of these ICI associated inflammatory toxicities across organ systems. Reviews include several in-depth overviews of tissue-specific irAE presentations. They address our current understanding of cellular and molecular drivers of irAEs and their relationship to anti-tumor immune response, including discussions on available preclinical models to study these mechanisms. They cover potential predictive biomarkers and genetic risk factors identified to-date, and discuss current management strategies being used, as well as clinical trials being developed using therapeutic candidates intended to limit irAE onset and improve patient outcomes. Collectively, these reviews summarize the state-of-the field of this evolving and complex subject, the critical challenges to ICI therapies posed by irAEs, along with the many opened questions and the strategies that should be the focus of future investigations.

The first set of six reviews delves deeply into our understanding of organ-specific irAEs. It is crucial to better understand the immunopathology of irAEs across organ-systems to allow for a more personalized immune suppression therapy and to spare negative effects on anti-tumor immunity. Although irAEs can target any organ system, toxicities occur more frequently at barrier tissues, including the gastrointestinal tract, skin, liver, and the lungs,15, 16, 26 and these are the focus of the first three reviews. The fact that barrier organs are so commonly involved suggests that the antigenic targets of the immune response may be the commensal microbiome, though this has not yet been demonstrated.

One of the first irAEs reported was vitiligo, which is a condition characterized by immune-mediated destruction of melanocytes that can occur in cancer patients with melanoma undergoing going treatment with various immunotherapies.15, 16, 26 Vitiligo is driven by T cells and antibodies targeting melanocyte antigens, which are also produced by melanoma. This immune response leads to the destruction of melanocytes, resulting in the characteristic depigmentation observed in affected individuals.28 Given these early observations, it was originally hypothesized that other irAEs might also result from targeting antigens common to healthy and malignant tissue, similarly to the mechanisms observed in vitiligo. Nonetheless, the fact that the majority of irAEs are not specific to a particular tumor type cast doubt on this hypothesis. Alternative hypotheses have now emerged that are discussed in the first three reviews. These posit a role for tissue-resident memory cells (TRM cells), which are long-lived memory effectors that reside in peripheral tissues and make up the majority of T cells in barrier tissue. TRM cells express PD-1 and other immune checkpoint molecules that control their activation. These cells play important roles in tissue surveillance and recall responses to pathogens.29-31 Recent studies have investigated whether ICI therapy triggers irAE by promoting the expansion and activation of local resident TRM cells. This work is discussed in the first three reviews covering irAE affecting the gastrointestinal track, the skin, and the lung.

The review by Michael Dougan provides a comprehensive review on the gastrointestinal (GI) toxicities caused by ICIs.32 Although most of the GI irAEs are mild in nature, their high occurrence rate makes them the leading cause of severe toxicities associated with ICIs. In his review, he provides an in-depth description of the clinical manifestations along with a thorough discussion of the immune mechanisms known to-date (e.g., gut CD8+ TRMs) associated with ICI-colitis, which is the most common GI toxicity associated with the use of ICIs.33 He also discusses risk factors associated with ICI-colitis, current standard of care, upcoming clinical trials, and the challenges related to the current lack of clinical trial data to guide management of patient care. Importantly, he highlights multiple unanswered questions in the field that require further investigation.

The second review from Blair Allais and colleagues is focused on dermatologic toxicities, which are the most frequently reported, the most visible, and among the earliest seen in cancer patients on ICI therapy.34 While most of these cutaneous immune-related adverse effects (cirAEs) are mild, severe presentations occur.35 Allais et al. provide a clear and thorough discussion of what is known about cirAEs through the different lenses being used to study them. These include harnessing the power of large datasets to understand clinical presentations and their association with outcomes. They establish specific definitions for each cirAEs, and their need for more targeted therapies. They describe leveraging cellular and molecular strategies to infer novel insights into disease mechanisms. They also describe the current understanding of immunosuppression treatment for managing cirAEs, as well as future opportunities in the field to help improve patient care.

Fiamma Berner and Lukas Flatz focused their review on two emerging classes of self-antigens involved in skin toxicity and pneumonitis in non-small cell lung cancer patients undergoing ICI therapy.36 They discuss how these self-antigen-specific T cells are believed to play a dual role by mediating irAE to skin and lung, and by promoting anti-tumor immune responses. Such dual association creates a challenge in distinguishing between the immune signature associated with irAE from those associated with treatment response. The review also highlights methods for the identification of self-antigen targets and discusses the role of antigens shared by organs affected by irAEs and tumors. Finally they discuss current challenges and solutions that may lead to successful cancer patient treatment with ICIs in the absence of problematic irAEs.

Gary Reynolds delves into rheumatological irAEs, which are disabling, can limit treatment efficacy and evolve to become chronic.37 These presentations can superficially share features of common immune-mediated rheumatic diseases (IMRDs),38 such as rheumatoid arthritis, psoriatic arthritis, Sjogren's syndrome, and polymyalgia rheumatica. This review discusses the parallels and differences between rheumatic irAEs and IMRDs, and highlight that, based on current knowledge, they are clinically and immunologically distinct entities.

Harish Seethapathy, Kavita Mistry, and Meghan Sise elegantly describe irAE manifestations in the kidney, which present as acute interstitial nephritis (ICI-AIN).39 They thoroughly explore the clinical manifestation of ICI-AIN and highlight its distinctive presentation compared to conventional drug-induced AIN. The authors provide a detailed description of current mechanistic understanding of ICI-AIN, and underscore the diagnostic and therapeutic implications that arise from these findings.

To conclude this first section of the issue, Jingyi Gong, Tomas Neilan, and Daniel A. Zlotoff present a thorough review on immune-related myocarditis (irMyocarditis), which, among all the cardiovascular toxicities, has been the most extensively studied due to its substantial risk of mortality.40 While irMyocarditis occurs in 1% of ICI recipients, the incidence is expected to continue rising with the increasing use of combination regimens to treat cancer patients.19-21, 41-43 It is fatal in 20%–40% of cases, which is approximately 10-fold higher than in non-ICI lymphocytic myocarditis.41-44 Nearly half of the cases result in major adverse cardiac events, such as severe arrhythmias, cardiogenic shock, and sudden cardiac death.41-44 Furthermore, the development of irMyocarditis is a near-absolute contraindication for future use of ICI therapy, which in turn may harm the oncologic prognosis.45 The authors frame their review around two primary objectives. First, they provide a comprehensive summary of the current understanding of irMyocarditis pathogenesis, and they compare it to other types of myocardial inflammation, which they examine across three categories: the cellular mediators, participating molecular signals and soluble factors, and T-cell receptor (TCR) clonality and specificity. Second, they discuss different hypotheses explaining its heightened morbidity compared to other forms of cardiac inflammation and why it only affects a limited number of cancer patients receiving ICI therapy. They conclude their elegant review by highlighting the many knowledge research and clinical gaps in the field requiring further investigation to enable developing better solutions for patient care.

While the above six reviews each focus on the clinical manifestations associated with specific organ system irAEs, the next section includes four reviews that delve more deeply into our biological understanding of irAE presentations across tissues, the known cellular drivers, their relationship to autoimmunity, and the available preclinical models that can be leveraged to study their mechanistic underpinnings. Importantly, defining the underlying disease mechanisms presents an opportunity not only to alleviate these detrimental side effects but to also improve the efficacy of ICI therapy itself.

Namrata Singh, Anne Hocking, and Jane Buckner provide an interesting review on the intersection between irAEs to ICI and autoimmune diseases.46 While several irAEs can phenocopy the clinical presentation of well-defined autoimmune diseases, it remains unclear whether these diseases are truly the same syndrome, especially as in many cases the ICI-associated presentation is more fulminate.47 This review explores whether the mechanisms responsible for inducing ICI-related irAEs are similar or different from those underlying spontaneous autoimmunity, focusing on ICI-diabetes, ICI-thyroiditis, and ICI-arthritis. The authors present a summary of our current understanding of the role of genetics and autoantibodies in the development of irAEs, and discusses the use of single-cell genomic strategies that have enabled the identification of specific T-cell signatures associated with irAEs. Ultimately, deciphering the relationship between these irAEs and spontaneous autoimmune diseases holds significant importance in terms of understanding basic mechanisms and clinical implications. Indeed, from a basic science standpoint, elucidating the connection between these clinical presentations will generate valuable insights into the fundamental mechanisms of autoimmunity and the biological principles of immune tolerance in human. From a clinical perspective, unraveling the relationship between irAEs and autoimmunity could help nominate therapeutic strategies for irAE based on therapies already clinically approved to mitigate autoimmunity.

Kavita Dhodapkar, Alyssa Duffy, and Madhav Dhodapkar discuss how the use of immune checkpoint inhibitor therapy can impact B-cell tolerance and drive alterations in the B-cell compartments.48 Their review explores the potential involvement of humoral immunity, and the role of specific B-cell subsets and autoantibodies in driving the irAE pathogenesis associated with the use of ICI. They also highlight the need to invest more resources in investigating the cross talk between T and B cells that drives the activation of pathogenic B cells, contributing to the development of irAEs.

Noah Earland and colleagues lay out a thorough perspective49 on their recently published work that analyzed a cohort of advanced melanoma using a multimodal strategy involving the use of single-cell RNA sequencing, as well as mass cytometry of peripheral blood cells to identify features predictive of irAE development.50 Their work showed that increased TCR diversity together with increased circulating levels of activated CD4 effector memory T cells are both associated with the development of severe irAEs, independent of the tissue affected.50 Through their review, they contextualize their results in comparison to other published work on irAE blood-based biomarkers. They examine possible mechanistic links between CD4 memory T cells and effector memory T cells, along with other factors that could contribute to irAE development, such as autoreactive tissue-resident memory T cells located at the tissue site of toxicity. Finally, they discuss factors that should be considered to empower future clinical translation of findings to develop and establish blood-based irAE-related clinical tests.

Presently, the pathophysiological mechanisms of irAE are poorly understood in part because preclinical mouse models underestimate the occurrence of irAEs compared to patient experience,51 or do not accurately replicate critical aspects of the human irAE biology.15, 16, 26, 52-54 Nevertheless, there is a multitude of available animal systems that could be tailored and leveraged to further model biological pathways identified in patients. For the last manuscript of this section, Morgan Cina, Jessica Venegas, and Arabella Young put together a very comprehensive review on the preclinical models available that capture some aspects of the irAE etiology across different organ systems. These can be used to study the biological underpinnings of irAEs, along with their relationship to anti-tumor immune responses.55 They examine the contexts for which these models are most useful, how they could be improved, as well as the advantages and disadvantages of different model systems. Approaches include in silico modeling of published datasets, in vitro organoid models, in vivo mouse and non-human primate models, as well as canine patients. They review why the development of model systems to study irAEs has not progressed rapidly. Particularly in comparison to efforts invested in establishing models to study therapeutic combination strategies. The authors end by discussing what the future holds for developing new preclinical models that capture with higher fidelity the breadth irAE presentations. Better models are critical to ultimately improve our mechanistic understanding of irAEs and their associated treatments.

The third and last section of this issue includes three reviews that examine what factors have been identified to-date that may help determine who will develop irAEs to ICI, whether we can predict which patient is predisposed to irAEs, and what is the relationship between irAE presentations and anti-tumor immune response. The ability to predict the occurrence of specific irAEs to ICI will be a critical step towards the development of prophylactic therapies and could prove valuable in making informed decisions regarding tailoring cancer treatment options for each patient.

One of the strategies used to identify disease predictors is to focus on uncovering germline susceptibility variants to ICI-related irAEs, which is elegantly reviewed in this issue by Alexander Gusev.56 Because these irAEs can phenocopy the clinical presentation of some sporadic autoimmune diseases, it has been hypothesized that they could be linked to autoimmunity through germline genetics and would be heritable.57-59 In this review, he summarizes current work mapping susceptibility germline variants to irAEs, outcome, and responses to ICIs. He also provides perspectives on recently published work from his group that leverages genome-wide association analysis to identify IL7 susceptibility variants associated with irAEs,60 findings that were subsequently independently replicated.61 He also discusses extant questions in the field, as well as opportunities to leverage germline genetic association studies to further our understanding of irAE etiology, in addition to improving our ability to predicting them.

There are currently no FDA approved biomarkers that predict the occurrence of irAEs, making it challenging to identify cancer patients for whom the risk of suffering from toxicity to ICI therapy is outweighed by the potential lifesaving benefits of receiving them. Moreover, the timing of these toxicities is not as predictable as side effects associated with other cancer treatments, such as chemotherapy and targeted therapy.62 Nevertheless, several efforts to map such biomarkers for irAE toxicities that could guide treatment selection, prevention, and monitoring have been initiated. These are thoroughly review by Rachel Goodman, Seungyeon Jung, Justin Balko, and Douglas Johnson.63 The authors describe different molecular and clinical biomarkers across three categories, including those that can help: (i) risk stratify patient pre-treatment to identify subjects likely to develop toxicities; (ii) diagnose irAEs, which remain challenging to do in many cases; and (iii) predict occurrence of irAEs during treatment course. These could enable tailoring clinical decision-making, including whether to alter treatment course or to increase the frequency of patient monitoring.

We conclude this issue with a comprehensive review by Steven M. Blum, Sherin Rouhani, and Ryan Sullivan on the molecular and clinical relationship between anti-tumor immune response and irAEs, along with a discussion of the role that irAE treatments may play in modulating the anti-tumor immune response.64 They examine the relationship between irAEs and anti-tumor immunity, which can be complex65 and influenced by factors such as the specific irAE-affected organ, tumor histology, and individual patient's characteristics. The authors highlight the importance of developing therapeutic strategies that can maximize the anti-tumor immunity efficacy of ICIs while minimizing the occurrence of irAEs by modulating both the host and the tumor immune environments, which will ultimately be essential to improve patient outcomes.

The landscape of cancer patient populations being treated with ICI therapies has significantly changed since their FDA approval for the treatment of metastatic melanoma in 2011; ICI is now used in indications with curative intent. For malignancies like metastatic melanoma, which were largely fatal before the introduction of ICI therapy, the benefits of ICIs far surpass the potential risks of developing irAEs in most patients, making such risk acceptable. In the context of adjuvant and neoadjuvant therapy, the risk-to-benefit ratio is less straightforward; while there has been improved disease-free survival, it is important to note that a considerable proportion of patients would be expected to achieve cancer remission without the use of ICIs, highlighting the need to reevaluate the irAEs risk versus the reward of being cancer-free. The illustration included in this introduction (Figure 1) captures the complicated balance between the benefits and risks associated with the use of ICI therapy to treat cancer patient. Despite the emergence of irAEs, it is important to remember that the introduction of ICI drugs has resulted in many good news stories, providing new hopes to cancer patients.

With the increasing utilization of ICI therapy among the cancer patient population, it is becoming increasingly critical to support research efforts aimed at understanding the root cause of irAE development, and how they can be mitigated while promoting anti-tumor immunity. Reviews included in this issue highlight how our understanding of irAEs has evolved over the years, along with the many challenges to be overcome and the remaining opened questions to be answered in the field, such as, which factors play a role in determining patient risk of experiencing irAEs? Can we predict which patient will develop irAEs? What factors contribute to the higher incidence of irAEs in certain organ systems? How are spontaneous autoimmune diseases and irAEs that phenocopy them related? Which factors influence the severity of an irAE in a particular patient, which can range from transient to chronic or fatal? How can we choose the most effective treatment strategies to treat irAEs while sparing the anti-tumor immune response? Once an irAE has improved upon treatment, which patient is at risk for redeveloping irAEs and which patient can be safely rechallenged with ICI therapy? Answering these outstanding questions will likely be the focus of upcoming studies in the immuno-oncology field.

Lastly, a broad theme that emerges from several reviews in this issue is the need to provide more financial support to this research across disciplines and to cross-institutional multidisciplinary teams spanning healthcare systems, academic researchers, federal research agencies, commercial partners, and patient advocacy groups to solve the underpinnings of irAEs and integrate critical discoveries into sustainable clinical solutions and applications. Even once irAE therapeutic targets are identified, the field will still necessitate cross-institutional collaborative endeavors to establish the clinical trial infrastructure required for evaluating these treatments. We owe it to cancer patients to work together as a community to find solutions to mitigate irAEs in order to improve their quality of life and enable them to optimally benefit from the life-saving virtues of ICI-therapy to fight their cancers.

ACV has been a paid consultant to Bristol Myers Squibb.

ACV is funded by the National Institute of Health Director's New Innovator Award (DP2CA247831), the Damon Runyon-Rachleff Innovation Award, The Melanoma Research Alliance Young Investigator Award (https://doi.org/10.48050/pc.gr.143739), the Massachusetts General Hospital (MGH) Transformative Scholar in Medicine Award, and the MGH Howard M. Goodman Fellowship.

Abstract Image

癌症患者免疫检查点免疫疗法后免疫相关不良事件的演变趋势
免疫检查点抑制剂(ICIs),包括靶向抗细胞毒性T淋巴细胞抗原4(CTLA-4)的抗体、抗程序性细胞死亡1(PD-1)、抗程序化细胞死亡1配体1(PD-L1),已经彻底改变了癌症治疗。1,2 PD-1和CTLA-4在T细胞激活时上调,并具有抑制外周T细胞反应的功能。这些受体维持免疫稳态,限制感染期间的组织损伤,并有助于预防自身免疫性疾病。CTLA-4通过细胞与CD28的外源性竞争来抑制T细胞活化,以结合相同的配体和细胞内在信号机制。3 PD-1:PD-:L1/L2途径还通过抑制TCR/CD28信号传导、靶细胞裂解和细胞因子产生来抑制T淋巴细胞反应。4 PD-1配体在不同的造血和非造血细胞中表达,表明其可能在外周组织中具有更大的影响,而CTLA-4及其配体主要在次级淋巴器官中表达。3总之,ICI免疫疗法利用后一种生物学来调节T细胞的激活和耐受性,使免疫系统克服癌症逃避免疫的问题,并重建其攻击肿瘤组织的能力。1,2 CTLA-4是20115-8年批准用于ICI治疗(即Ipilimumab)的第一个靶点,用于治疗晚期黑色素瘤。从那时起,美国食品药品监督管理局(FDA)批准了8种靶向PD-1/PD-L1或CTLA-4途径的药物,用于80种适应症中的20多种癌症类型。9重要的是,ICI在佐剂环境中的使用也在增加,导致接受ICI治疗的患者数量进一步增加。接受ICIs治疗的癌症患者现在预计寿命更长;但是长期暴露于ICI治疗也使他们面临与该系列治疗相关的长期并发症的更大风险。10-14不幸的是,ICIs的成功受到治疗诱导的炎症毒性(称为免疫相关不良事件)的出现的限制。irAE可以影响身体的任何器官系统,其严重程度各不相同,导致患者的严重发病率和医疗系统的巨大成本。这些irAE可在治疗期间或治疗后发生,通常需要免疫抑制治疗或停止ICI治疗,从而限制了ICI治疗的潜在救生益处。1、2、15、16根据治疗方案(即药物类型、组合和剂量),66%-96%的患者发生irAE,7%-59%的患者经历严重irAE。17,18暴发性和致命性irAE——最常见的是心肌炎/肌炎、肺炎和神经事件(如格林-巴利综合征和重症肌无力)——约0.4%的癌症患者接受抗PD-1/PD-L1单药治疗,1.2%的患者接受PD-1/CTLA-4联合治疗,但它们往往在最初的3 ICI.15启动后的几个月,16免疫肿瘤学仍然是一个快速发展的领域,CTLA-4和PD-1之外的更多共抑制途径可能在不久的将来成为有价值的治疗靶点。例如,relatlimab,一种阻断淋巴细胞抗原3(LAG-3)的化合物,最近已被批准与抗PD1疗法联合使用。21该靶点主要存在于衰竭的T细胞中,可能解决与肿瘤微环境相关的T细胞相关的无能。随着ICIs在未来几年成为更多肿瘤类型的一线和二线癌症治疗,以及ICIs在辅助治疗和联合治疗中的使用增加,以提高抗肿瘤反应疗效并克服治疗耐药性,22,23预计irAE的发生率将继续上升,并严重限制ICIs的救生潜力。有趣的是,在ICI治疗环境中发生的irAE与特发性自身免疫性疾病非常相似,包括类风湿性关节炎(RA)、自身免疫性肝炎和炎症性肠病。1此外,PD-1和CTLA-4的多态性与各种自身免疫性疾病有关,如1型糖尿病、甲状腺炎、Graves病、乳糜泻,系统性红斑狼疮和类风湿性关节炎15,16,24,25——观察到的与治疗相关的irAE可以具有共同的临床特征。irAE的治疗方法与自身免疫性疾病相同,包括类固醇、生物制剂,有时还包括手术。因此,对irAE发病机制的深入了解,在这种情况下,确切的疾病触发和疾病发生的时间是已知的,不仅可能对癌症患者的护理有影响,而且可能对我们对特发性自身免疫性疾病的理解有影响。 研究与irAE相关的细胞和相关分子模式,可以为了解患者自身免疫系统开始对正常组织产生炎症反应的确切时刻提供一个难得的窗口。这样的研究还可以对全身免疫调节机制产生新的见解,并可以进一步了解CTLA-4和PD-1如何维持免疫耐受。26因此,解读irAE的发病机制不仅可能对癌症患者的护理有意义,而且还可能为驱动和维持自发性自身免疫性疾病的最早步骤提供重要的见解,这些疾病在完全确定后很难识别和研究。虽然我们可以假设irAE发作的触发因素是T细胞信号通路的操纵和组织特异性免疫耐受的破坏,1,2,15,16但对驱动和维持针对正常组织的过度免疫激活的下游机制仍知之甚少。这在一定程度上是由于临床前动物模型尚未概括出irAE临床表现的全谱。15,16迄今为止提出的疾病机制的例子包括肿瘤和受影响组织之间的共享抗原、先前存在的亚临床炎症、先前存在自身抗体、抗体依赖性细胞毒性和环境绝缘。15,27鉴于irAE临床表现的异质性和广度,影响不同器官系统的irAE可能具有不同的病理驱动因素。在特定组织内具有相似组织学特征的irAE中,多种机制内型也可能导致类似的临床表现。我们对ICI的irAE机制的了解有限,严重限制了靶向治疗方案的开发。迄今为止,irAE的治疗管理指南几乎完全是基于小型回顾性临床研究和专家意见制定的。由于不同器官系统的毒性和严重程度的驱动机制可能不同,因此需要医学界对每个irAE单独进行前瞻性治疗试验,而不是将多个irAE一起研究。在过去几年中,在理解ICI相关irAE的生物学驱动因素和制定有效的缓解策略方面进展有限。对irAE的细胞和分子基础的进一步研究对于确定更好的毒性预测因子以预防其发生,以及开发新的、靶向的和改进的治疗方案至关重要,这些解决方案可以减轻这些毒性,同时保持抗肿瘤免疫反应。本期包括该领域顶尖科学家和医学专家的13篇综述文章,他们讨论了我们目前对这些ICI相关的跨器官系统炎症毒性的理解。综述包括对组织特异性irAE表现的几个深入综述。它们阐述了我们目前对irAE的细胞和分子驱动因素及其与抗肿瘤免疫反应的关系的理解,包括对研究这些机制的可用临床前模型的讨论。它们涵盖了迄今为止确定的潜在预测性生物标志物和遗传风险因素,并讨论了目前正在使用的管理策略,以及正在使用旨在限制irAE发作和改善患者预后的候选治疗药物进行的临床试验。总之,这些综述总结了这一不断发展和复杂主题的领域现状,irAE对ICI疗法提出的关键挑战,以及许多悬而未决的问题和应成为未来研究重点的策略。第一组六篇综述深入探讨了我们对器官特异性irAE的理解。至关重要的是,要更好地了解跨器官系统的irAE的免疫病理学,以便进行更个性化的免疫抑制治疗,并避免对抗肿瘤免疫产生负面影响。尽管irAE可以靶向任何器官系统,但毒性更频繁地发生在屏障组织,包括胃肠道、皮肤、肝脏和肺部,15、16、26,这些是前三篇综述的重点。屏障器官如此普遍地参与这一事实表明,免疫反应的抗原靶点可能是共生微生物组,尽管这一点尚未得到证实。最先报道的irAE之一是白癜风,这是一种以免疫介导的黑色素细胞破坏为特征的疾病,可发生在接受各种免疫疗法治疗的癌症黑色素瘤患者中。15,16,26白癜风是由T细胞和靶向黑色素细胞抗原的抗体驱动的,这些抗原也是由黑色素瘤产生的。 这种免疫反应会导致黑色素细胞的破坏,导致在受影响的个体中观察到的特征性色素脱失。28鉴于这些早期观察结果,最初假设其他irAE也可能是靶向健康和恶性组织常见的抗原引起的,类似于在白癜风中观测到的机制。尽管如此,大多数irAE并不是特定肿瘤类型的特异性,这一事实使人们对这一假设产生了怀疑。在前三篇综述中讨论的替代假设现在已经出现。这些为组织驻留记忆细胞(TRM细胞)发挥作用,TRM细胞是驻留在外周组织中的长寿命记忆效应器,占屏障组织中T细胞的大部分。TRM细胞表达PD-1和其他控制其激活的免疫检查点分子。这些细胞在组织监测和对病原体的回忆反应中发挥着重要作用。29-31最近的研究调查了ICI治疗是否通过促进局部驻留TRM细胞的扩增和激活来触发irAE。这项工作在前三篇综述中进行了讨论,涵盖了影响胃肠道、皮肤和肺部的irAE。Michael Dougan的综述对ICIs引起的胃肠道(GI)毒性进行了全面综述。32尽管大多数胃肠道irAE性质温和,但其高发生率使其成为与ICIs相关的严重毒性的主要原因。在他的综述中,他对临床表现进行了深入描述,并对迄今为止已知的与ICI结肠炎相关的免疫机制(如肠道CD8+TRM)进行了彻底讨论,ICI结肠炎是与使用ICI相关的最常见的胃肠道毒性。33他还讨论了与ICI肠炎相关的风险因素、当前的护理标准、即将进行的临床试验,以及与目前缺乏指导患者护理管理的临床试验数据有关的挑战。重要的是,他强调了该领域中需要进一步调查的多个未回答的问题。Blair Allais及其同事的第二篇综述集中在皮肤毒性方面,这是最常见、最明显的,也是在接受ICI治疗的癌症患者中最早发现的,出现了严重的表现。35 Allais等人通过研究cirAE的不同视角,对cirAE进行了清晰而彻底的讨论。其中包括利用大型数据集的力量来理解临床表现及其与结果的关系。他们为每种cirAE建立了具体的定义,以及他们对更有针对性的治疗的需求。他们描述了利用细胞和分子策略来推断对疾病机制的新见解。他们还描述了目前对免疫抑制治疗治疗cirAE的理解,以及该领域未来帮助改善患者护理的机会。Fiamma Berner和Lukas Flatz将他们的综述集中在两类新出现的自身抗原上,这两类自身抗原与接受ICI治疗的癌症非小细胞肺癌患者的皮肤毒性和肺炎有关。36他们讨论了这些自身抗原特异性T细胞如何被认为通过介导对皮肤和肺部的irAE和促进抗肿瘤免疫反应发挥双重作用。这种双重关联在区分与irAE相关的免疫特征和与治疗反应相关的免疫标志方面带来了挑战。该综述还强调了识别自身抗原靶点的方法,并讨论了受irAE和肿瘤影响的器官共享抗原的作用。最后,他们讨论了在没有问题irAE的情况下,可能导致癌症患者成功接受ICI治疗的当前挑战和解决方案。Gary Reynolds深入研究了风湿病学irAE,这些疾病具有致残性,可能限制治疗效果,并发展为慢性疾病,38,如类风湿性关节炎、银屑病关节炎、干燥综合征和风湿性多肌痛。这篇综述讨论了风湿性irAE和IMRD之间的异同,并强调,根据目前的知识,它们在临床和免疫学上是不同的实体。Harish Seethapathy、Kavita Mistry和Meghan Sise优雅地描述了肾脏中的irAE表现,表现为急性间质性肾炎(ICI-AIN)。39他们深入探讨了ICI-AIN的临床表现,并强调了其与传统药物诱导的AIN相比的独特表现。作者详细描述了目前对ICI-AIN的机制理解,并强调了这些发现所带来的诊断和治疗意义。最后,龚静怡、托马斯·内兰和丹尼尔A。 兹洛托夫对免疫相关心肌炎(irMyocarditis)进行了全面的回顾,在所有心血管毒性中,由于其巨大的死亡风险,该病已被研究得最为广泛。40虽然1%的ICI受试者发生irmyocartis,但随着癌症患者联合治疗方案的使用增加,预计发病率将继续上升。19-21,41-43它在20%-40%的病例中是致命的,比非ICI淋巴细胞性心肌炎高出约10倍。41-44近一半的病例会导致严重的心脏不良事件,如严重心律失常、心源性休克和心源性猝死。41-44此外,发展成心肌炎是未来使用ICI治疗的几乎绝对禁忌症,这反过来可能会损害肿瘤学的预后。45作者围绕两个主要目标进行综述。首先,他们对目前对病毒性心肌炎发病机制的理解进行了全面总结,并将其与其他类型的心肌炎症进行了比较,他们对其进行了三类检查:细胞介质、参与分子信号和可溶性因子,以及T细胞受体(TCR)的克隆性和特异性。其次,他们讨论了不同的假设,解释了与其他形式的心脏炎症相比,其发病率升高的原因,以及为什么它只影响少数接受ICI治疗的癌症患者。他们在总结其优雅的综述时强调了该领域的许多知识研究和临床空白,需要进一步调查,以开发更好的患者护理解决方案。虽然以上六篇综述都集中在与特定器官系统irAE相关的临床表现上,但下一节包括四篇综述,更深入地研究我们对组织间irAE表现的生物学理解、已知的细胞驱动因素、它们与自身免疫的关系,以及可用于研究其机制基础的可用临床前模型。重要的是,确定潜在的疾病机制不仅有机会减轻这些有害副作用,而且有机会提高ICI治疗本身的疗效。Namrata Singh、Anne Hocking和Jane Buckner对irAE与ICI和自身免疫性疾病之间的交叉点进行了有趣的综述,特别是在许多情况下,ICI相关的表现更具暴发性。47本综述探讨了诱导ICI相关irAE的机制与潜在的自发自身免疫机制是否相似或不同,重点关注ICI糖尿病、ICI甲状腺炎和ICI关节炎。作者总结了我们目前对遗传和自身抗体在irAE发展中的作用的理解,并讨论了单细胞基因组策略的使用,这些策略能够识别与irAE相关的特异性T细胞特征。最终,破译这些irAE与自发性自身免疫性疾病之间的关系对于理解基本机制和临床意义具有重要意义。事实上,从基础科学的角度来看,阐明这些临床表现之间的联系将对自身免疫的基本机制和人类免疫耐受的生物学原理产生有价值的见解。从临床角度来看,揭示irAE和自身免疫之间的关系有助于根据临床上已经批准的减轻自身免疫的疗法,为irAE提出治疗策略。Kavita Dhodapkar、Alyssa Duffy和Madhav Dhodapkal讨论了免疫检查点抑制剂治疗的使用如何影响B细胞耐受性并驱动B细胞区的改变。48他们的综述探讨了体液免疫的潜在参与,以及特定B细胞亚群和自身抗体在驱动与ICI使用相关的irAE发病机制中的作用。他们还强调,需要投入更多的资源来研究T细胞和B细胞之间的串扰,对irAE的发展做出了贡献。Noah Earland及其同事对他们最近发表的工作进行了全面的展望49,该工作使用涉及单细胞RNA测序的多模式策略分析了一组晚期黑色素瘤,以及外周血细胞的质谱细胞术,以确定预测irAE发展的特征。50他们的工作表明,TCR多样性的增加以及活化的CD4效应记忆T细胞循环水平的增加都与严重irAE的发展有关,与受影响的组织无关。 50通过他们的综述,他们将自己的结果与其他已发表的基于irAE血液的生物标志物的工作进行了比较。他们研究了CD4记忆T细胞和效应记忆T细胞之间可能的机制联系,以及可能导致irAE发展的其他因素,如位于毒性组织部位的自身反应组织驻留记忆T细胞。最后,他们讨论了应该考虑的因素,以使研究结果的未来临床翻译能够开发和建立基于血液的irAE相关临床测试。目前,irAE的病理生理机制尚不清楚,部分原因是与患者经验相比,临床前小鼠模型低估了irAE的发生,51或者没有准确复制人类irAE生物学的关键方面。15,16,26,52-54然而,有许多可用的动物系统可以被定制和利用,以进一步模拟在患者中识别的生物途径。在本节的最后一份手稿中,Morgan Cina、Jessica Venegas和Arabella Young对现有的临床前模型进行了非常全面的综述,这些模型捕捉了不同器官系统中irAE病因的某些方面。这些可用于研究irAE的生物学基础,以及它们与抗肿瘤免疫反应的关系。55他们研究了这些模型最有用的背景,如何改进它们,以及不同模型系统的优缺点。方法包括对已发表的数据集、体外类器官模型、体内小鼠和非人灵长类动物模型以及犬类患者进行计算机建模。他们回顾了为什么研究irAE的模型系统的开发进展不快。特别是与建立研究治疗组合策略的模型的努力相比。作者最后讨论了开发新的临床前模型的未来,这些模型可以更逼真地捕捉irAE的广度。更好的模型对于最终提高我们对irAE及其相关治疗的机制理解至关重要。本期的第三部分也是最后一部分包括三篇综述,研究迄今为止已经确定的哪些因素可能有助于确定谁会发展为ICI的irAE,我们是否可以预测哪些患者易患irAE,以及irAE表现与抗肿瘤免疫反应之间的关系。预测ICI特异性irAE发生的能力将是开发预防性疗法的关键一步,并可能被证明有助于就为每位患者量身定制癌症治疗方案做出明智的决定。用于确定疾病预测因子的策略之一是专注于揭示ICI相关irAE的种系易感性变体,Alexander Gusev在本期文章中对此进行了优雅的综述。56因为这些irAE可以表型复制一些散发性自身免疫性疾病的临床表现,据推测,它们可能通过种系遗传学与自身免疫有关,并且是可遗传的。57-59在这篇综述中,他总结了目前绘制IRAE易感性种系变体、结果和对ICIs的反应的工作。他还对其团队最近发表的工作提供了看法,该工作利用全基因组关联分析来识别与irAE相关的IL7易感性变体,60项发现随后被独立复制。61他还讨论了该领域现存的问题,以及利用种系遗传关联研究来进一步了解irAE病因的机会,以及提高我们预测它们的能力。目前没有FDA批准的生物标志物可以预测irAE的发生,这使得识别癌症患者的挑战性,因为接受ICI治疗的潜在救生益处超过了他们遭受ICI治疗毒性的风险。此外,这些毒性的发生时间不如其他癌症治疗(如化疗和靶向治疗)相关的副作用可预测。62然而,已经开始了几项绘制irAE毒性生物标志物的工作,这些生物标志物可以指导治疗选择、预防和监测。Rachel Goodman、Seungyeon Jung、Justin Balko和Douglas Johns对此进行了全面审查。63作者描述了三类不同的分子和临床生物标志物,包括那些有帮助的生物标志物:(i)对患者治疗前进行风险分层,以确定可能产生毒性的受试者;(ii)诊断irAE,这在许多情况下仍然具有挑战性;和(iii)预测治疗过程中irAE的发生。这些措施可以调整临床决策,包括是否改变疗程或增加患者监测频率。 我们以Steven M.Blum、Sherin Rouhani和Ryan Sullivan对抗肿瘤免疫反应和irAE之间的分子和临床关系的全面综述来结束这个问题,并讨论了irAE治疗在调节抗肿瘤免疫应答中可能发挥的作用。64他们研究了irAE和抗肿瘤免疫之间的关系,其可能是复杂的65,并受到诸如受特定irAE影响的器官、肿瘤组织学和个体患者特征等因素的影响。作者强调了开发治疗策略的重要性,该策略可以通过调节宿主和肿瘤免疫环境,最大限度地提高ICIs的抗肿瘤免疫效力,同时最大限度地减少irAE的发生,这最终对改善患者的预后至关重要。自2011年美国食品药品监督管理局批准治疗转移性黑色素瘤以来,接受ICI疗法治疗的癌症患者群体的情况发生了显著变化;ICI现在用于具有治疗意图的适应症。对于转移性黑色素瘤等恶性肿瘤,在采用ICI治疗之前基本上是致命的,ICI的益处远远超过大多数患者发生irAE的潜在风险,使这种风险可以接受。在辅助治疗和新辅助治疗的情况下,风险收益比不那么直接;尽管无病生存率有所提高,但值得注意的是,在不使用ICIs的情况下,相当大比例的患者有望获得癌症缓解,这突出表明需要重新评估irAE的风险与无癌回报。本简介中的插图(图1)反映了使用ICI治疗癌症患者的益处和风险之间的复杂平衡。尽管出现了irAE,但重要的是要记住,ICI药物的引入带来了许多好消息,为癌症患者带来了新的希望。随着ICI疗法在癌症患者群体中的使用率不断提高,支持旨在了解irAE发展的根本原因以及如何在促进抗肿瘤免疫的同时减轻其影响的研究工作变得越来越重要。本期综述强调了多年来我们对irAE的理解是如何演变的,以及需要克服的许多挑战和该领域需要回答的悬而未决的问题,例如,哪些因素在确定患者经历irAE的风险中发挥作用?我们能预测哪些患者会出现irAE吗?哪些因素导致某些器官系统中irAE的发病率较高?自发性自身免疫性疾病和表型分析的irAE有何关联?哪些因素会影响特定患者的irAE严重程度,从短暂到慢性或致命?我们如何选择最有效的治疗策略来治疗irAE,同时保留抗肿瘤免疫反应?一旦irAE在治疗后得到改善,哪个患者有再次发生irAE的风险,哪个患者可以安全地接受ICI治疗?回答这些悬而未决的问题可能是免疫肿瘤学领域即将进行的研究的重点。最后,从本期的几篇综述中得出的一个广泛主题是,需要为这项跨学科的研究提供更多的财政支持,并为医疗系统、学术研究人员、联邦研究机构、商业合作伙伴、,以及患者倡导团体,以解决irAE的基础,并将关键发现整合到可持续的临床解决方案和应用中。即使确定了irAE治疗靶点,该领域仍需要跨机构合作,以建立评估这些治疗所需的临床试验基础设施。我们有责任让癌症患者作为一个社区共同努力,找到减轻irAE的解决方案,以提高他们的生活质量,并使他们能够从ICI疗法的拯救生命的优点中最佳受益,从而对抗癌症。ACV一直是百时美施贵宝的付费顾问。ACV由美国国家卫生研究所主任新创新者奖(DP2CA247831)、Damon Runyon Rachleff创新奖、黑色素瘤研究联盟青年研究员奖资助(https://doi.org/10.48050/pc.gr.143739)、马萨诸塞州总医院(MGH)医学变革学者奖和MGH霍华德·M·古德曼奖学金。
本文章由计算机程序翻译,如有差异,请以英文原文为准。
求助全文
约1分钟内获得全文 求助全文
来源期刊
Immunological Reviews
Immunological Reviews 医学-免疫学
CiteScore
16.20
自引率
1.10%
发文量
118
审稿时长
4-8 weeks
期刊介绍: Immunological Reviews is a specialized journal that focuses on various aspects of immunological research. It encompasses a wide range of topics, such as clinical immunology, experimental immunology, and investigations related to allergy and the immune system. The journal follows a unique approach where each volume is dedicated solely to a specific area of immunological research. However, collectively, these volumes aim to offer an extensive and up-to-date overview of the latest advancements in basic immunology and their practical implications in clinical settings.
×
引用
GB/T 7714-2015
复制
MLA
复制
APA
复制
导出至
BibTeX EndNote RefMan NoteFirst NoteExpress
×
提示
您的信息不完整,为了账户安全,请先补充。
现在去补充
×
提示
您因"违规操作"
具体请查看互助需知
我知道了
×
提示
确定
请完成安全验证×
copy
已复制链接
快去分享给好友吧!
我知道了
右上角分享
点击右上角分享
0
联系我们:info@booksci.cn Book学术提供免费学术资源搜索服务,方便国内外学者检索中英文文献。致力于提供最便捷和优质的服务体验。 Copyright © 2023 布克学术 All rights reserved.
京ICP备2023020795号-1
ghs 京公网安备 11010802042870号
Book学术文献互助
Book学术文献互助群
群 号:481959085
Book学术官方微信