Skip to main content
NIHPA Author Manuscripts logoLink to NIHPA Author Manuscripts
. Author manuscript; available in PMC: 2021 Mar 17.
Published in final edited form as: Curr Opin Immunol. 2019 Sep 23;61:54–59. doi: 10.1016/j.coi.2019.08.004

Autoimmune complications of cancer immunotherapy

Kavita M Dhodapkar 1
PMCID: PMC7968474  NIHMSID: NIHMS1678714  PMID: 31557690

Abstract

Immunotherapy of cancer with blockade of inhibitory immune checkpoints and adoptive cell therapies have led to impressive clinical responses in several cancers. However, with increasing utilization of these therapies, immune-related adverse events have emerged as a major obstacle. Herein I discuss recent insights into the immunobiology of these toxicities. Deeper understanding of the underlying pathogenic mechanisms, cellular and molecular pathways involved, similarities and differences with spontaneous autoimmunity, and identification of clinically relevant predictive biomarkers is needed to develop optimal approaches to prevent and treat these toxicities, without compromising the therapeutic benefit from these immune therapies. These events may also provide a unique window into mechanisms underlying spontaneous autoimmunity.

The clinical challenge of immune-related adverse events (irAEs) in cancer immunotherapy

Immune-therapies such as blockade of immune checkpoints and adoptive cell therapies have begun to transform the therapeutic landscape in cancer. In particular, antibodies targeting inhibitory immune checkpoint (ICP) molecules cytotoxic T-lymphocyte-associated protein-4 (CTLA-4), programmed death-1 (PD-1), and programmed death ligand-1 (PD-L1), either alone or in combination, can lead to durable tumor regressions and have now been approved for treatment of several different cancers. However, use of these therapies has also been associated with increase in irAEs [1,2]. This is particularly evident in the setting of combination therapy with CTLA-4 and PD-1 blockade where increase in tumor regressions following combination therapy is associated with a marked increase in Grade 3 and 4 irAEs resulting in significant morbidity and discontinuation of therapy in up to one third of patients [2,3••]. Therefore, irAEs represent a major therapeutic challenge, not only in terms of the morbidities associated with these toxicities, but also limiting the ability to administer ICPs. The pattern, severity and onset of specific irAEs seems to differ based on the specific agent, or combination [2]. For example, CTLA-4 blockade led to colitis and hypophysitis, while anti-PD-1 therapy led to higher rates of pneumonitis [4]. Combination checkpoint blockade (CCB) with anti-CTLA-4 and anti-PD-1 is associated with high rates of early and multi-organ autoimmunity, including hepatitis [3••]. These clinical events raise several questions. For example, a) to what degree the ICP-induced irAEs resemble conventional autoimmune diseases, and represent activation of an occult autoimmune state, or do they represent alternate mechanisms of auto-inflammation initiated by altered tolerance mechanisms?; b) what are the mechanisms that impact involvement of specific tissues; c) what are the key cell types that mediate these effects and do they differ from those responsible for anti-tumor effects; d) does the biology of ‘early’ irAEs which occur within days of starting therapy differ from those manifestations that develop over a more delayed course such as several months or years after initiating therapy; d) how can we identify patients at risk and effectively prevent such complications without impacting tumor regression? Deeper understanding of the underlying mechanisms, particularly in patients undergoing these therapies is therefore urgently needed.

Insights from animal models: opportunities and limitations

CTLA-4 deficient mice rapidly succumb to lymphoproliferative disease leading to multi-organ failure [5]. In contrast, PD-1 deficient mice have a milder phenotype with delayed and tissue-restricted autoimmunity in a strain-dependent manner [6]. Autoimmunity is also evident for mice deficient in other immune checkpoints now entering the clinic, although preclinical data have not been entirely replicated in the clinic. For example, combined loss of PD-1 and LAG-3 results in lethal autoimmunity in mice, although in early clinical experience, the safety profile of anti-LAG3 plus nivolumab was similar to that of nivolumab alone [7]. While these models were critical to the early discoveries relating to immune-regulatory function of these molecules, they also do not seem to faithfully recapitulate the patterns of irAEs observed in the clinic, such as pneumonitis in patients treated with anti-PD-1 therapy [8]. Part of the limitation may be due to the fact that many of the cancer animal models rely on B6 or BALB/c strains which are generally resistant to autoimmune complications. Species-specific differences in biology may also represent a challenge. For example, deficiency of regulatory T cells (Tregs) is a more prominent feature of CTLA-4 blockade in mouse models than in human trials [9]. Studies in autoimmunity-prone strains may also be target-specific. For example, in autoimmune diabetes-prone non-obese diabetic (NOD) mice, anti-PD-1 rapidly induces diabetes [10], while the effects of anti-CTLA-4 are mostly restricted to newborn mice [11]. The risk of autoimmunity may also depend on the underlying genetic makeup of the individual. Recent advances in genetic humanization of mice may allow dissection of these effects by studying effects of checkpoint blockade in vivo utilizing human samples [12]. Nonetheless, these studies point to the unmet need to systematically study patients receiving immune checkpoint therapies with a particular emphasis on dissecting mechanisms of autoimmunity, as well the development of new models that faithfully recapitulate autoimmune toxicities of these agents in the clinic (Figure 1).

Figure 1.

Figure 1

Pathways to autoimmunity following checkpoint blockade.

Several mechanisms have been proposed for induction of irAEs that involve pathogenic B or T cells. Panel (a). Patients with pre-existing autoreactive T cells against antigens expressed in involved tissues may undergo activation following ICP blockade and contribute to autoimmunity; Panel (b). Autoimmunity may be mediated by activation and expansion of preexisting autoreactive B cell clones such as CD21lo B cells. These clones and downstream plasmablasts may be responsible for the generation of pathogenic Igs and may also serve as antigen-presenting cells to activate T cells; Panel (c): Activation of autoreactive B cells may depend on help provided by TFH or TPH cells. The antigens targeted by pathogenic B or T cells in most patients are currently unknown and in some instances they may be cross-reactive to antigens expressed on tumor cells.

Natural autoimmunity versus irAEs

Both irAEs and natural autoimmunity can potentially share several features including autoantibodies, autoreactive T cells and underlying genetic risk factors, but may differ in terms of underlying pathogenesis, even in the setting of tissue-specific autoimmunity. Antibodies against islet-associated antigens are present in over 90% of patients with type I diabetes but in less than half of the patients who develop this as a complication of ICP blockade [13]. Evaluation of global antibody profiles in patients receiving ICP-blockade identified signatures associated with patients who developed severe irAEs [14].Patients with spontaneous autoimmunity such as lupus have distinct molecular signatures in B cells leading to altered B cell differentiation and function [15,16]. It remains to be established whether identification of pre-existing autoimmune signatures will identify patients at greater risk for irAEs. Implications of underlying genetic susceptibility risks may also exist but may differ between natural autoimmunity and irAE and need further study. For example, HLA markers DQ8 and DR53 are associated with spontaneous hypophysitis but not ICP-induced hypophysitis [17]. Patients who experienced ICP-induced diabetes had a significantly higher proportion of HLA-DR4, exceeding that observed in Caucasians with spontaneous type I diabetes [13]. Certain autoimmune disease such as lupus are more common in African-American patients, but their susceptibility to ICP-induced irAEs needs further study [18]. As the development of irAEs in the clinic is a relatively recent phenomenon, long term consequences of such autoimmunity have not yet been studied. ICP-induced irAEs in principle offer the potential for fundamental insights into the early origins of autoimmunity because the biology of such early clinical phase of natural autoimmunity is very hard to study in humans. Deeper understanding of similarities (and differences) between natural and ICP-induced autoimmunity including dissection of antigenic targets and cellular mechanisms may help identify patients at increased risk for the development of irAEs, which in turn will aid optimal management of these patients.

Cellular and pathophysiologic mechanisms

Both B and T cells have been implicated in the pathogenesis of autoimmune states. Histo-pathologic analyses of involved tissuessuchascolitisinthecontextofanti-CTLA-4-induced autoimmunity was found to show enrichment of both B and T cells [19]. Humans with CTLA-4 deficiency have increased propensity for the development of autoimmunity, and exhibit alterations in both B and T cells [20]. In melanoma patients receiving combination ICP blockade, patients who experienced early changes in B cells were at increased risk for the development of autoimmune implications [21••]. These changes involved a decline in circulating B cells, concurrent with enrichment of CD21lo subset of B cells and plasmablasts. These changes were associated with an increase in plasma CXCL13 as a biomarker of germinal center activity [21••,22]. Early decline in B cells as well as pre-existing anti-acetylcholine receptors antibodies were also associated with high risk of irAEs in patients with thymoma treated with immune checkpoint blockade [23]. CD21lo double negative-2 (DN2) cells are now appreciated as a distinct subset enriched in autoreactive B cells and an increase in this B cell subset has been described in some patients with autoimmune diseases [2430]. CD21lo B cells were initially thought to be anergic or exhausted B cells [26]. However, a recent study reported that these cells are potent antigen presenting cells [31]. They lack lymphoid homing chemokines and instead express chemokine receptors important for trafficking to sites of inflammation in non-lymphoid tissue [31]. CD21lo B cells were also recently described as new germinal center emigrants primed for plasma cell differentiation [32] that show overlap with tissue-homing innate-like B cells [33]. Recent studies in lupus patients have characterized components of the human EF B cell activation pathway including activated naı¨ve B cells that differentiate into effector B cells (DN2 cells), representing epigenetically primed precursors of ASC [15,16,34,35•,36,37]. Together these studies point to the extrafollicular pathway as a major contributor to disease-specific autoantibodies in lupus, which is induced by TLR7 and a combination of IFNγ and IFNλ stimulation. Recent data also suggest that CD21lo B cells are the precise subset of human B cells specifically enriched for PD-1 expression [21••] and undergo in vivo activation and proliferation following CCB therapy positioning them as important targets of CCB therapy. Combination blockade in humans may therefore essentially phenocopy the features of germline CTLA4 deficient cohorts with increase in CD21lo B cells. Additional blockade of PD1 may then provide release of inhibition on PD1-expressing CD21lo B cells, ultimately leading to B cell mediated autoimmune complications.This subset of B cells is therefore an attractive candidate as a target for mediating irAEs following ICP blockade and improved characterization of this subset of pathogenic B cells may lead to novel therapeutic approaches targeting these cells.

Prior studies have also tried to correlate alterations in T cell repertoire in patients receiving ICP blockade and the risk of irAEs. For example, an increase in circulating T cell clones was linked to the risk of severe irAEs following ipilimumab therapy [38•]. In another study, immune toxicities followingCTLA-4blockadewerealso associated with early diversification of T cell repertoire [39]. At present, the relationship, if any, between T cells mediating anti-tumor effects versus those that might contribute to irAEs remains unclear. While T cells infiltrating or resident in tumors are emerging as key players in the context of tumor immunity [40,41], their relationship to those infiltrating organs/tissues involved in autoimmune inflammation needs to be clarified. Such studies will require tissue biopsies of involved organs as well as tumor tissue.

The antigenic targets of pathogenic antibodies causing autoimmunity or T cells infiltrating involved organs remains an area of active exploration. In particular, it would be of interest to determine if some of these antibodies also target antigens expressed on tumor cells. Another possibility is that these irAEs represent activation of clinically occult autoimmunity and involves preexisting autoreactive antibodies or T cells. Further studies of systematic dissection of antigenic targets of auto-reactive B or T cells should help clarify these issues. A recent study has shown that patients with metastatic but non-progressive tumors have persistent B cell responses expressing shared paratopes targeting public tumor antigens [42]. Another mechanism involved in the pathogenesis of irAEs may involve direct binding of the therapeutic antibody to the involved tissue. For example, in the setting of anti-CTLA-4-associated hypophysitis, the hypophyseal cells were shown to express CTLA-4, which led to direct antibody binding [43••]. Similarly, islet cells in pancreas were shown to express PD-L1, which may permit direct binding of the antibody to these targets [44]. It is important to note that some patients may experience autoimmune manifestations involving multiple organs following ICP blockade. In this setting, several pathogenic mechanisms may coexist at the same time in different tissues. Differences in underlying pathogenic mechanisms may even apply to toxicities within the same tissue. For example, dermatologic manifestations of ICP-blockade are diverse and range from mild maculopapular rash to bullous lesions, vitiligo, lichenoid reactions and psoriatic lesions [45,46]. Differences in pathogenesis may also underlie the kinetics of irAEs and may differ between those that occur early versus those that occur many months after initiating ICP blockade. Finally, it is now well appreciated that each of the current ICP blockade strategies leads to a distinct genomic signature in humans [47]. This is also likely for many of the novel combination therapies that are currently being tested in the clinic. These differences may account for the differing rates and patterns of irAEs observed in the clinic following these therapies. In the setting of combination ICP therapy, the risk of irAE may depend more on one drug versus the other [48].For example, the risk of early irAE following combination of anti-CTLA-4 and anti-PD-1 seems to depend more on the dose of anti-CTLA-4 than anti-PD-1.

Extrinsic factors impacting irAEs

In addition to underlying host genetics, preexisting autoreactivity or occult autoimmune disease, there is also increasing interest in the possibility that extrinsic influences may impact the risk of irAEs and thus may be a modifiable risk factor. One example of such extrinsic factors may be the nature of signals from the microbiome. In preclinical models, certain species of Bacteroides and Burkholderiales not only potentiated anti-tumor effects of CTLA-4 blockade but also reduced the risk of colitis [49,50•]. In melanoma patients receiving checkpoint blockade, the presence of Firmicutes species was enriched in patients experiencing colitis [51]. At present it remains to be established if the observed effect is mostly related to involved tissues (e.g. gut flora and risk of colitis) or if it impacts systemic immunity as well. The mechanisms underlying these observations remain to be clarified and may include cross-reactivity or mimicry of endogenous molecules or functional alterations in immune cells. Nonetheless, if prospectively confirmed in independent studies, alteration of microbial composition may represent a novel approach to modify risk of irAEs following ICP blockade.

Opportunities for therapeutic intervention or prevention

Studies evaluating tumor regressions in patients experiencing irAEs have yielded conflicting results, with some reporting no impact [52] while others reporting improvement in response [4]. As corticosteroids are the mainstay of therapy of these toxicities, an important consideration is whether it will also impact anti-tumor efficacy. Several studies suggest that corticosteroid usage does not seem to impair outcome. However, this does not exclude the possibility that steroids may have blunted a favorable outcome. Appreciation of dysregulation of cytokines such as tumor necrosis factor (TNF) and IL-17 has led to application of anti-cytokine approaches in the treatment of some of these toxicities [53]. Some examples include tumor necrosis factor-targeted therapies in steroid refractory colitis and the use of interleukin-6 targeted approaches for therapy of cytokine release syndrome following infusion of chimeric-antigen receptor T cells [54]. Recent insights that early changes in B cells predict the risk of autoimmune complications following combination ICP blockade [21••] have prompted initiation of randomized clinical studies specifically targeting B cells. B cells may in principle serve both a favorable and unfavorable role in tumor immunity [55,56]. Several subsets of B cells including those expressing PD1, have been shown to promote tumor progression in preclinical models [5759]. Selective depletion of B cells in patients undergoing combination ICP blockade may therefore help dissect the role of B cells and determine if this approach can reduce irAEs and enhance antitumor efficacy. It is reassuring to note that depletion of B cells does not seem to impact anti-tumor effects of ICP blockade in preclinical models of melanoma [60]. It is however through systematic evaluation of patients undergoing irAE-targeted therapies that we will identify patient-specific biomarkers and targets to prevent these complications. These autoimmune phenomena may also provide a unique window into understanding basic mechanisms of immune tolerance and early events into the pathogenesis of spontaneous autoimmunity.

Acknowledgements

KMD is supported in part by funds from N.I.H. (NIH1R01CA238471-01), Cure Foundation and Melanoma Innovation Fund.

Footnotes

Conflict of interest statement

Nothing declared.

References and recommended reading

Papers of particular interest, published within the period of review, have been highlighted as

• of special interest

•• of outstanding interest

  • 1.June CH, Warshauer JT, Bluestone JA: Is autoimmunity the Achilles’ heel of cancer immunotherapy? Nat Med 2017, 23:540–547. [DOI] [PubMed] [Google Scholar]
  • 2.Boutros C, Tarhini A, Routier E, Lambotte O, Ladurie FL, Carbonnel F et al. : Safety profiles of anti-CTLA-4 and anti-PD-1 antibodies alone and in combination. Nat Rev Clin Oncol 2016, 13:473–486. [DOI] [PubMed] [Google Scholar]
  • 3.••.Sznol M, Ferrucci PF, Hogg D, Atkins MB, Wolter P, Guidoboni M et al. : Pooled analysis safety profile of Nivolumab and Ipilimumab combination therapy in patients with advanced melanoma. J Clin Oncol 2017. JCO2016721167. [DOI] [PubMed] [Google Scholar]; An important report describing the clinical patterrns of autoimmune complications following combination checkpoint blockade.
  • 4.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, 35:785–792. [DOI] [PubMed] [Google Scholar]
  • 5.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, 270:985–988. [DOI] [PubMed] [Google Scholar]
  • 6.Nishimura H, Okazaki T, Tanaka Y, Nakatani K, Hara M, Matsumori A et al. : Autoimmune dilated cardiomyopathy in PD-1 receptor-deficient mice. Science 2001, 291:319–322. [DOI] [PubMed] [Google Scholar]
  • 7.Okazaki T, Okazaki IM, Wang J, Sugiura D, Nakaki F, Yoshida T et al. : PD-1 and LAG-3 inhibitory co-receptors act synergistically to prevent autoimmunity in mice. J Exp Med 2011, 208:395–407. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 8.Naidoo J, Wang X, Woo KM, Iyriboz T, Halpenny D, Cunningham J et al. : Pneumonitis in patients treated with anti-programmed death-1/programmed death ligand 1 therapy. J Clin Oncol 2017, 35:709–717. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 9.Arce Vargas F, Furness AJS, Litchfield K, Joshi K, Rosenthal R, Ghorani E et al. : Fc effector function contributes to the activity of human anti-CTLA-4 antibodies. Cancer Cell 2018, 33:649–663 e4. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 10.Ansari MJ, Salama AD, Chitnis T, Smith RN, Yagita H, Akiba H et al. : The programmed death-1 (PD-1) pathway regulates autoimmune diabetes in nonobese diabetic (NOD) mice. J Exp Med 2003, 198:63–69. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 11.Luhder F, Hoglund P, Allison JP, Benoist C, Mathis D: Cytotoxic T lymphocyte-associated antigen 4 (CTLA-4) regulates the unfolding of autoimmune diabetes. J Exp Med 1998, 187:427–432. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 12.Das R, Strowig T, Verma R, Koduru S, Hafemann A, Hopf S et al. : Microenvironment-dependent growth of preneoplastic and malignant plasma cells in humanized mice. Nat Med 2016, 22:1351–1357. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 13.Stamatouli AM, Quandt Z, Perdigoto AL, Clark PL, Kluger H, Weiss SA et al. : Collateral damage: insulin-dependent diabetes induced with checkpoint inhibitors. Diabetes 2018, 67:1471–1480. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 14.Gowen MF, Giles KM, Simpson D, Tchack J, Zhou H, Moran U et al. : Baseline antibody profiles predict toxicity in melanoma patients treated with immune checkpoint inhibitors. J Transl Med 2018, 16:82. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 15.Jenks SA, Cashman KS, Woodruff MC, Lee FE, Sanz I: Extrafollicular responses in humans and SLE. Immunol Rev 2019, 288:136–148. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 16.Tipton CM, Fucile CF, Darce J, Chida A, Ichikawa T, Gregoretti I et al. : Diversity, cellular origin and autoreactivity of antibody-secreting cell population expansions in acute systemic lupus erythematosus. Nat Immunol 2015, 16:755–765. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 17.Heaney AP, Sumerel B, Rajalingam R, Bergsneider M, Yong WH, Liau LM: HLA markers DQ8 and DR53 are associated with lymphocytic hypophysitis and may aid in differential diagnosis. J Clin Endocrinol Metab 2015, 100:4092–4097. [DOI] [PubMed] [Google Scholar]
  • 18.Tiu AC, Potdar R, Djibo DA, Masab M, Dourado C: Clinical outcomes of African American patients with advanced or metastatic non-small cell lung cancer on Nivolumab in a single community-based cancer center. Med Oncol 2018, 35:109. [DOI] [PubMed] [Google Scholar]
  • 19.Beck KE, Blansfield JA, Tran KQ, Feldman AL, Hughes MS, Royal RE et al. : Enterocolitis in patients with cancer after antibody blockade of cytotoxic T-lymphocyte-associated antigen 4. J Clin Oncol 2006, 24:2283–2289. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 20.Kuehn HS, Ouyang W, Lo B, Deenick EK, Niemela JE, Avery DT et al. : Immune dysregulation in human subjects with heterozygous germline mutations in CTLA4. Science 2014, 345:1623–1627. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 21.••.Das R, Bar N, Ferreira M, Newman AM, Zhang L, Bailur JK et al. : Early B cell changes predict autoimmunity following combination immune checkpoint blockade. J Clin Invest 2018, 128:715–720. [DOI] [PMC free article] [PubMed] [Google Scholar]; Early change in B cells with an increase in CD21lo B cell subset associated with increased risk of irAEs. One of the first studies linking these B cells to checkpoint-induced irAEs.
  • 22.Havenar-Daughton C, Lindqvist M, Heit A, Wu JE, Reiss SM, Kendric K et al. : CXCL13 is a plasma biomarker of germinal center activity. Proc Natl Acad Sci U S A 2016, 113:2702–2707. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 23.Mammen AL, Rajan A, Pak K, Lehky T, Casciola-Rosen L, Donahue RN et al. : Pre-existing antiacetylcholine receptor autoantibodies and B cell lymphopaenia are associated with the development of myositis in patients with thymoma treated with avelumab, an immune checkpoint inhibitor targeting programmed death-ligand 1. Ann Rheum Dis 2019, 78:150–152. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 24.Thorarinsdottir K, Camponeschi A, Gjertsson I, Martensson IL: CD21-/low B cells: a snapshot of a unique B cell subset in health and disease. Scand J Immunol 2015, 82:254–261. [DOI] [PubMed] [Google Scholar]
  • 25.Thorarinsdottir K, Camponeschi A, Cavallini N, Grimsholm O, Jacobsson L, Gjertsson I et al. : CD21(-/low) B cells in human blood are memory cells. Clin Exp Immunol 2016, 185:252–262. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 26.Isnardi I, Ng YS, Menard L, Meyers G, Saadoun D, Srdanovic I et al. : Complement receptor 2/CD21- human naive B cells contain mostly autoreactive unresponsive clones. Blood 2010, 115:5026–5036. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 27.Wehr C, Eibel H, Masilamani M, Illges H, Schlesier M, Peter HH et al. : A new CD21low B cell population in the peripheral blood of patients with SLE. Clin Immunol 2004, 113:161–171. [DOI] [PubMed] [Google Scholar]
  • 28.Warnatz K, Wehr C, Drager R, Schmidt S, Eibel H, Schlesier M et al. : Expansion of CD19(hi)CD21(lo/neg) B cells in common variable immunodeficiency (CVID) patients with autoimmune cytopenia. Immunobiology 2002, 206:502–513. [DOI] [PubMed] [Google Scholar]
  • 29.Saadoun D, Terrier B, Bannock J, Vazquez T, Massad C, Kang I et al. : Expansion of autoreactive unresponsive CD21-/low B cells in Sjogren’s syndrome-associated lymphoproliferation. Arthritis Rheum 2013, 65:1085–1096. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 30.Terrier B, Joly F, Vazquez T, Benech P, Rosenzwajg M, Carpentier W et al. : Expansion of functionally anergic CD21-/low marginal zone-like B cell clones in hepatitis C virus infection-related autoimmunity. J Immunol 2011, 187:6550–6563. [DOI] [PubMed] [Google Scholar]
  • 31.Shimabukuro-Vornhagen A, Garcia-Marquez M, Fischer RN, Iltgen-Breburda J, Fiedler A, Wennhold K et al. : Antigen-presenting human B cells are expanded in inflammatory conditions. J Leukoc Biol 2017, 101:577–587. [DOI] [PubMed] [Google Scholar]
  • 32.Lau D, Lan LY, Andrews SF, Henry C, Rojas KT, Neu KE et al. : Low CD21 expression defines a population of recent germinal center graduates primed for plasma cell differentiation. Sci Immunol 2017, 2. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 33.Rakhmanov M, Gutenberger S, Keller B, Schlesier M, Peter HH, Warnatz K: CD21low B cells in common variable immunodeficiency do not show defects in receptor editing, but resemble tissue-like memory B cells. Blood 2010, 116:3682–3683. [DOI] [PubMed] [Google Scholar]
  • 34.Jenks SA, Cashman KS, Woodruff MC, Lee FE-H, Sanz I: Extrafollicular responses in humans and SLE. Immunol Rev 2019, 288:136–148. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 35.•.Jenks SA, Cashman KS, Zumaquero E, Marigorta UM, Patel AV, Wang X et al. : Distinct effector B cells induced by unregulated toll-like receptor 7 contribute to pathogenic responses in systemic lupus erythematosus. Immunity 2018, 49:725–739.e6. [DOI] [PMC free article] [PubMed] [Google Scholar]; An important study characterizing the properties of CD21lo B cells in lupus.
  • 36.Zumaquero E, Stone SL, Scharer CD, Jenks SA, Nellore A, Mousseau B et al. : IFNgamma induces epigenetic programming of human T-bet(hi) B cells and promotesTLR7/8 and IL-21 induced differentiation. eLife 2019, 8. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 37.Scharer CD, Blalock EL, Barwick BG, Haines RR, Wei C, Sanz I et al. : ATAC-seq on biobanked specimens defines a unique chromatin accessibility structure in naive SLE B cells. Sci Rep 2016, 6:27030. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 38.•.Subudhi SK, Aparicio A, Gao J, Zurita AJ, Araujo JC, Logothetis CJ et al. : Clonal expansion of CD8 T cells in the systemic circulation precedes development of ipilimumab-induced toxicities. Proc Natl Acad Sci U S A 2016, 113:11919–11924. [DOI] [PMC free article] [PubMed] [Google Scholar]; An early study linking changes in T cells with the risk of irAEs.
  • 39.Oh DY, Cham J, Zhang L, Fong G, Kwek SS, Klinger M et al. : Immune toxicities elicted by CTLA-4 blockade in cancer patients are associated with early diversification of the T-cell repertoire. Cancer Res 2017, 77:1322–1330. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 40.Boddupalli CS, Bar N, Kadaveru K, Krauthammer M, Pornputtapong N, Mai Z et al. : Interlesional diversity of T cell receptors in melanoma with immune checkpoints enriched in tissue-resident memory T cells. JCI Insight 2016, 1:e88955. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 41.Dhodapkar KM: Role of tissue-resident memory in intra-tumor heterogeneity and response to immune checkpoint blockade. Front Immunol 2018, 9:1655. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 42.DeFalco J, Harbell M, Manning-Bog A, Baia G, Scholz A, Millare B et al. : Non-progressing cancer patients have persistent B cell responses expressing shared antibody paratopes that target public tumor antigens. Clin Immunol 2018, 187:37–45. [DOI] [PubMed] [Google Scholar]
  • 43.••.Iwama S, De Remigis A, Callahan MK, Slovin SF, Wolchok JD, Caturegli P: Pituitary expression of CTLA-4 mediates hypophysitis secondary to administration of CTLA-4 blocking antibody. Sci Transl Med 2014, 6 230ra. [DOI] [PubMed] [Google Scholar]; Induction of hypophysitis linked to the expression of CTLA4 on pituatory cells.
  • 44.Rui J, Deng S, Arazi A, Perdigoto AL, Liu Z, Herold KC: beta Cells that resist immunological attack develop during progression of autoimmune diabetes in NOD mice. Cell Metab 2017, 25:727–738. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 45.Sibaud V: Dermatologic reactions to immune checkpoint inhibitors: skin toxicities and immunotherapy. Am J Clin Dermatol 2018, 19:345–361. [DOI] [PubMed] [Google Scholar]
  • 46.Naidoo J, Schindler K, Querfeld C, Busam K, Cunningham J, Page DB et al. : Autoimmune bullous skin disorders with immune checkpoint inhibitors targeting PD-1 and PD-L1. Cancer Immunol Res 2016, 4:383–389. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 47.Das R, Verma R, Sznol M, Boddupalli CS, Gettinger SN, Kluger H et al. : Combination therapy with anti-CTLA-4 and anti-PD-1 leads to distinct immunologic changes in vivo. J Immunol 2015, 194:950–959. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 48.Coutzac C, Adam J, Soularue E, Collins M, Racine A, Mussini C et al. : Colon immune-related adverse events: anti-CTLA-4 and anti-PD-1 blockade induce distinct immunopathological entities. J Crohns Colitis 2017, 11:1238–1246. [DOI] [PubMed] [Google Scholar]
  • 49.Vetizou M, Pitt JM, Daillere R, Lepage P, Waldschmitt N, Flament C et al. : Anticancer immunotherapy by CTLA-4 blockade relies on the gut microbiota. Science 2015, 350:1079–1084. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 50.•.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. [DOI] [PMC free article] [PubMed] [Google Scholar]; An important study linking gut microbiota to risk of irAEs.
  • 51.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, 28:1368–1379. [DOI] [PubMed] [Google Scholar]
  • 52.Horvat TZ, Adel NG, Dang TO, Momtaz P, Postow MA, Callahan MK et al. : Immune-related adverse events, need for systemic immunosuppression, and effects on survival and time to treatment failure in patients with melanoma treated with Ipilimumab at memorial Sloan Kettering cancer center. J Clin Oncol 2015, 33:3193–3198. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 53.Tarhini AA, Zahoor H, Lin Y, Malhotra U, Sander C, Butterfield LH et al. : Baseline circulating IL-17 predicts toxicity while TGF-beta1 and IL-10 are prognostic of relapse in ipilimumab neoadjuvant therapy of melanoma. J Immunother Cancer 2015, 3:39. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 54.Teachey DT, Lacey SF, Shaw PA, Melenhorst JJ, Maude SL, Frey N et al. : Identification of predictive biomarkers for cytokine release syndrome after chimeric antigen receptor T-cell therapy for acute lymphoblastic leukemia. Cancer Discov 2016, 6:664–679. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 55.Gunderson AJ, Coussens LM: B cells and their mediators as targets for therapy in solid tumors. Exp Cell Res 2013, 319:1644–1649. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 56.Liudahl SM, Coussens LM: B cells as biomarkers: predicting immune checkpoint therapy adverse events. J Clin Invest 2018, 128:577–579. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 57.Xiao X, Lao XM, Chen MM, Liu RX, Wei Y, Ouyang FZ et al. : PD-1hi identifies a novel regulatory B-cell population in human hepatoma that promotes disease progression. Cancer Discov 2016, 6:546–559. [DOI] [PubMed] [Google Scholar]
  • 58.Pylayeva-Gupta Y, Das S, Handler JS, Hajdu CH, Coffre M, Koralov SB et al. : IL35-producing B cells promote the development of pancreatic neoplasia. Cancer Discov 2016, 6:247–255. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 59.Affara NI, Ruffell B, Medler TR, Gunderson AJ, Johansson M, Bornstein S et al. : B cells regulate macrophage phenotype and response to chemotherapy in squamous carcinomas. Cancer Cell 2014, 25:809–821. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 60.Damsky W, Jilaveanu L, Turner N, Perry C, Zito C, Tomayko M et al. : B cell depletion or absence does not impede anti-tumor activity of PD-1 inhibitors. J Immunothe Cancer 2019, 7:153. [DOI] [PMC free article] [PubMed] [Google Scholar]

RESOURCES