Next Article in Journal
Monitoring SARS-CoV-2 Surrogate TGEV Individual Virions Structure Survival under Harsh Physicochemical Environments
Next Article in Special Issue
Immune Microenvironment of Muscular-Invasive Urothelial Carcinoma: The Link to Tumor Immune Cycle and Prognosis
Previous Article in Journal
Reovirus Activated Cell Death Pathways
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Immune Cells in Hyperprogressive Disease under Immune Checkpoint-Based Immunotherapy

1
School of Medicine, Tsinghua University, Haidian District, Beijing 100084, China
2
Hepatopancreatbiliary Center, Tsinghua University Affiliated Beijing Tsinghua Changgung Hospital, Changping District, Beijing 102218, China
*
Author to whom correspondence should be addressed.
Cells 2022, 11(11), 1758; https://doi.org/10.3390/cells11111758
Submission received: 11 March 2022 / Revised: 18 May 2022 / Accepted: 23 May 2022 / Published: 27 May 2022
(This article belongs to the Special Issue Tumor Immune Microenvironment for Effective Therapy)

Abstract

:
Immunotherapy, an antitumor therapy designed to activate antitumor immune responses to eliminate tumor cells, has been deeply studied and widely applied in recent years. Immune checkpoint inhibitors (ICIs) are capable of preventing the immune responses from being turned off before tumor cells are eliminated. ICIs have been demonstrated to be one of the most effective and promising tumor treatments and significantly improve the survival of patients with multiple tumor types. However, low effective rates and frequent atypical responses observed in clinical practice limit their clinical applications. Hyperprogressive disease (HPD) is an unexpected phenomenon observed in immune checkpoint-based immunotherapy and is a challenge facing clinicians and patients alike. Patients who experience HPD not only cannot benefit from immunotherapy, but also experience rapid tumor progression. However, the mechanisms of HPD remain unclear and controversial. This review summarized current findings from cell experiments, animal studies, retrospective studies, and case reports, focusing on the relationships between various immune cells and HPD and providing important insights for understanding the pathogenesis of HPD.

1. Introduction

Immunotherapy has emerged as a new and developing method of tumor treatment in recent years and is now widely used in clinical practice [1]. A total of 10 immune checkpoint inhibitors (ICIs) targeting programmed cell death protein 1 (PD-1) or its ligand programmed cell death ligand 1 (PD-L1), such as pembrolizumab, nivolumab, and atezolizumab, as well as one ICI targeting cytotoxic T-lymphocyte-associated antigen 4 (CTLA-4), ipilimumab, have been approved for clinical application in the United States and China [2]. However, many studies have reported that the reactivities of patients treated with ICIs differ significantly. Firstly, a long duration of tumor shrinkage occurs in 10% to 30% of patients treated with ICIs, which is much higher than that achieved with other tumor treatments, and overall survival (OS) is improved unprecedentedly [3]. Secondly, under ICI treatment, some patients exhibit mixed responses or pseudoprogression, which is defined as an initial surge in the tumor burden, followed by tumor shrinkage [4]. Finally, 4% to 29% of patients cannot benefit from ICIs. Instead, they experience faster and more aggressive tumor progression than expected, which is known as hyperprogressive disease (HPD) [5]. As shown in Table 1, the correlations between HPD and ICIs have been shown across multiple types of tumor. A number of studies have proved the influence of HPD on OS, progression-free survival (PFS), and the rate of tumor progression [6,7,8,9,10,11,12,13,14,15,16,17,18,19,20,21,22,23,24,25]. As ICIs become more prevalent, improved knowledge of HPD is urgently needed to precisely determine which patients should receive immunotherapy. MDM2 and EGFR mutations have only been considered as potential risk factors, rather than causal factors, because these gene mutations predispose tumors to progression. Thus, current studies are committed to determining the exact cause of HPD [26]. In addition, ICI-based immunotherapy is designed to restore immune cell attack against tumors by blocking PD-1 or CTLA-4. Thus, immune cells certainly play a crucial role in the development of HPD. As more data become available, the reasons behind this phenomenon may be uncovered, allowing the application of preemptive measures to mitigate HPD. The purpose of this review is to provide an update about HPD and explain the possible mechanisms by which ICIs induce HPD by influencing various types of immune cells.

2. CD8+ T Cells

The immune checkpoint blockade (ICB) was originally designed to restore T-cell attack against tumor cells by blocking the primary negative regulator of T-cell function, PD-1, with ICIs [27]. Therefore, the responses of tumor patients to ICIs are influenced by the infiltration of activated CD8+ T cells into the tumors. Tumors filled with T cells are generally considered to be more sensitive to ICIs, while tumors lacking tumor-infiltrating lymphocytes (TILs) have a low absent response to ICIs [1]. Because CD8+ T cells play a crucial role in immunotherapy, we speculate that CD8+ T cells may also contribute to the development of HPD.

2.1. Exhausted CD8+ T (Tex) Cells

In patients with chronic infection and tumors, long-term exposure to persistent antigens and inflammation causes continuous CD8+ T cell stimulation to the point that the cells gradually lose effector function and memory T-cell characteristics, which is a condition known as T-cell exhaustion [28]. Functionally different from effector T cells and memory T cells, Tex cells are characterized by effector function loss, continuously increased regulation of inhibitory receptor (IR) expression, changes in epigenetic and transcriptional profiles, and changes in metabolic patterns [29]. T-cell exhaustion is one of the main factors of immune dysfunction in cancer patients [30]. In multiple tumors, Tex cells exhibit dysfunction in producing effector cytokines, limiting the immune response to tumor cells [31,32,33,34].
It has been reported that PD-1CD8+ T cells lack terminal differentiation and proliferative ability and are driven into an exhausted state [35]. Kim et al. [11] demonstrated that a lower number of CCR7CD45RA T cells, which are effector or memory cells, combined with a higher number of TIGIT+ T cells, which are severely depleted tumor-specific CD8+ T cells, was associated with the development of HPD and poor survival in tumor patients. However, Wartewig et al. [36] observed that anti-PD-1/PD-L1 treatment could induce HPD in T-cell non-Hodgkin’s lymphoma mice. Researchers demonstrated that PD-1 inhibited carcinogenic signaling by increasing the level of PTEN while decreasing PI3K/AKT and NF-κB signaling pathway activity. Hence, anti-PD-1 antibodies are able to rapidly accelerate T-cell growth. Ratner et al. [37] reported HPD after nivolumab treatment in three patients with chronic, smoldering acute lymphoma and adult T-cell leukemia.
Stein et al. [38] analyzed the correlation between TILs and tumor cells. Since T cells that infiltrated into breast tumors showed low expression of granzyme B [39] and high expression of PD-1 [40], they focused on cognate nonlytic antigen-specific interactions between CD8+ T cells and tumor cells. They proved that CD8+ T cells were activated by and interacted with breast cancer cells but could not kill them, thereby inducing the expression of pluripotency-associated genes in tumor cells and thus inducing cancer stem cell (CSC) formation [41,42]. They further demonstrated that T cells deprived of cytotoxic activity could reshape tumor cells after symbiotic interaction with CSCs. They hypothesized that ICIs could promote these nonlytic interactions and lead to hyperprogression through the dedifferentiation of tumor cells. This mechanism is the basis of the epigenetic resistance hypothesis, suggesting that CSCs contribute to HPD, but the associated biological mechanism remains to be discovered.
It is worth mentioning that Jiang et al. [43] introduced homeostasis of the activated and exhausted CD8+ T cells in tumor microenvironment (TME). They explained the curative effect of ICIs via the expansion and differentiation of progenitor Tex cells and believed that the depletion of the T cell progenitors and the effector function of Tex cells were the causes of the ineffectiveness of immune monotherapy under exorbitant tumor burden. Therefore, they proposed that ICIs should be combined with vaccines, chemotherapy, radiotherapy, targeted therapy, and cytokines to reduce tumor burden and expand the progenitor T cell pool.

2.2. Immune Checkpoint Compensation Mechanisms

PD-1 blockade triggers compensation mechanisms that suppress CD8+ T-cell activity [44,45,46]. This reverse regulatory feedback may counteract the benefits of anti-PD-1 treatment based on T-cell activation and in turn establish an enhanced immunosuppressive TME, laying the foundation for the progression of HPD [47]. Huang et al. [44] reported that TILs isolated from ovarian cancer-model mice simultaneously expressed multiple ICI receptors, such as PD-1, CTLA-4, and lymphocyte-activation gene 3 (LAG-3), which are involved in the creation of a highly immunosuppressive TME. Interestingly, PD-1 blockade confers compensatory enhanced expression of LAG-3 and CTLA-4 in CD8+ T cells, negating the benefits of ICB. Koyama et al. [48] analyzed the TME in mice with lung adenocarcinoma and in two non-small-cell lung cancer (NSCLC) patients treated with anti-PD-1 agents. They observed T-cell immunoglobulin and mucin domain-3 (TIM3) overexpression in tumor-permeable cytotoxic CD8+ T cells after immunotherapy failure. TIM3 expression level was significantly related to the duration of PD-1 blockade. These results suggest that anti-PD-1/PD-L1 antibodies activate compensatory immunosuppression and immune escape processes and may lead to HPD. Thus, the combined application of multiple ICIs may be more effective in inhibiting tumor progression than anti-PD-1 monotherapy. However, the mechanisms of interactions between different immune checkpoints need to be further explored.

2.3. IL-10/IL-10R

Sun et al. [49] reported that circulating tumor-specific CD8+ T cells in advanced melanoma with high PD-1 expression also have high expression levels of IL-10R and that the level of IL-10R expression can be further increased by PD-1/PD-L1 blockade, making these T cells more sensitive to IL-10. Therefore, the PD-1 blockade-induced secretion of IL-10 by innate immune cells, such as monocytes and the upregulation of IL-10R expression in T cells, may lead to severe inhibition of the antitumor activity of CD8+ T cells, thus forming a vicious cycle of immune escape that is conducive to tumor growth. In addition, the researchers also reported that the inhibition of IL-10 enhanced the role of anti-PD-1 antibodies in amplifying tumor-specific CD8+ T cells, thereby enhancing their antitumor activity. These findings further prove that IL-10 and IL-10R are involved in the development of HPD.

3. CD4+ T Cells

Immunotherapy research has often focused on CD8+ T cells because of their ability to eliminate tumor cells. However, CD4+ T cells have attracted attention in the field because they are not only crucial for promoting CD8+ T cell functions, preventing CD8+ T cell depletion or inducing CD8+ T cell memory, but also able to directly or indirectly kill tumor cells [50].

3.1. Regulatory T (Treg) Cells

In addition to effector cells, the T lymphocyte family includes an immunomodulatory subgroup called Treg cells, whose role is to negatively regulate other immune cells, prevent the overactivation of the immune response, and play a role in a wide range of diseases, such as allergies, chronic infections, and parasitic infections [51]. However, the presence of Treg cells is disadvantageous to hosts with tumors because they limit an effective antitumor immune response. Kamada et al. [52] reported that the proportions of effector regulatory T (eTreg) cells/CD8+ T cells, Ki67+ Treg cells/Ki67+CD8+ T cells, and Ki67+ Treg cells decreased significantly in non-HPD patients after treatment with anti-PD-1 antibodies, while these proportions in HPD patients remained stable or even increased slightly. This finding suggested that if the number of CD8+ T cells is insufficient to overcome Treg cells, the possibility of HPD development is greatly increased. Furthermore, Treg cells have also been shown to express immune checkpoints, such as PD-1; thus, Treg cells can also be targeted by anti-PD-1 agents [53]. Researchers have observed that knocking out PD-1 in Treg cells or blocking PD-1 with monoclonal antibodies (mAbs) caused Treg cells to gain a stronger proliferative ability and a stronger immunosuppression ability, thus leading to a stronger ability to promote tumor growth. This finding suggested that PD-1+ Treg cells play a key role in anti-PD-1 treatment-mediated HPD in advanced gastric cancer. In addition, Ratner et al. [37,54] demonstrated that nivolumab led to rapid progression in patients with adult T-cell leukemia/lymphoma (ATLL). They identified a novel relationship between tumor-resident Tregs and ATLL cells and revealed the tumor suppressive effect of PD-1 in ATLL.
Furthermore, in Treg cells treated with PD-1 blockade, the expression of immune checkpoints is upregulated, and the immunosuppressive function is enhanced. Thus, the antitumor immunity of some patients after anti-PD-1 treatment is not enhanced but greatly weakened, which leads to the occurrence of HPD. Interestingly, CTLA-4 was found to be strongly expressed in Treg cells [55]. Anti-CTLA-4 treatment increased the presence of Ki67+ Treg cells [52]. Furthermore, the combination of anti-CTLA-4 antibodies and anti-PD-1 antibodies was associated with a lower incidence of HPD than other ICI combinations, and CTLA-4, OX-40, or CCR4-targeted therapy might be a strategy for preventing HPD through Treg cell consumption [56]. In addition, selective PD-1/PD-L1 inhibition may lead to tumor immune evasion and accelerate tumor growth by increasing the number of Treg cells infiltrating and circulating in the tumor [57].

3.2. Other Subsets of CD4+ T Cells

We should consider that other CD4+ T cells beyond Treg cells are equally important. CD4+CD28 T cells are a cell subpopulation with unique biological effects that frequently appear in some autoimmune diseases [58]. Due to their lack of CD28, which is necessary for a cell-specific immune response and the most important costimulatory molecule on the T-cell surface, these unique cells not only have abnormal immune function but also have the characteristics of autoreactivity, massive expansion, and a long lifespan [59]. Arasanz et al. [60] found that CD4+CD28 T cells in the peripheral blood of lung cancer patients with HPD were amplified after PD-1 treatment, and high tumor growth dynamics scores were associated with the presence of CD4+CD28 T-cell subsets in patients with HPD.
In addition, Zappasodi et al. [61] observed in melanoma mice that a subset of CD4+Foxp3PD-1high T cells can perform immunosuppressive functions similar to those of Treg cells, but their RNA expression levels may be more similar to those of follicular helper T (Tfh) cells. Interestingly, while anti-PD-1 treatment reduced the numbers of these cells, anti-CTLA4 treatment increased their intratumoral abundance. This result suggests that this cell subpopulation could also respond to ICB, proliferate under anti-CTLA-4 treatment, and acquire negative regulatory immune properties, which might contribute to the development of HPD (Figure 1).

3.3. Exhausted CD4+ T Cells

Another potential mechanism of HPD is the correlation between exhausted CD4+ T cells and anti-PD-1 treatment. The current understanding of CD4+ T cell exhaustion is obviously insufficient. However, the negative effects of CD4+ T cell exhaustion on proliferation, cytokine production, B-cell help, and CD8+ effector functions have been reported. Furthermore, exhausted CD4+ T cells upregulate immune-regulatory proteins, such as TIM3 and PD-1, paralleling phenotypes observed in exhausted CD8+ T cells [62]. Unlike non-HPD patients, HPD patients showed abnormal dilation of peripheral exhausted memory CD4+ T cells after the initial administration of anti-PD-1/PD-L1 antibodies [60]. Arasanz et al. [60] monitored peripheral blood mononuclear cells (PBMCs) in NSCLC patients treated with anti-PD-1/PD-L1 antibodies, and peripheral exhausted CD4+ T-cell proliferation was observed in patients with HPD. They proposed that the rapid expansion of peripheral CD28CD4+ T cells is an early distinguishing feature of ICIs-induced HPD in NSCLC. Although the role of exhausted CD4+ T cells is not fully understood, these studies provide important evidence that these cells might also contribute to the progression of HPD.

3.4. IFN-γ

While IFN-γ is considered to be a key factor in antitumor immunity [63,64], Xiao et al. [65] demonstrated that IFN-γ could promote immune escape and papilloma development by enhancing a Th17-associated inflammatory reaction. Thus, IFN-γ can promote either antitumor immunity or immune escape according to the pathological background and the level of selective stress [66]. Sakai et al. [67] reported that in a mouse model of Mycobacterium tuberculosis infection, PD-1 led to the extensive penetration of CD4+ T cells into the lung parenchyma and the production of large amounts of IFN-γ, causing rapid disease progression, compared with that observed in wild-type mice. In addition, mutations in genes encoding IFN-γ signaling pathway components, such as IFN-γ receptor and JAK1/2, have been identified as potential mechanisms of resistance against anti-PD-1/PD-L1 and anti-CTLA-4 antibodies [64,68]. Champiat et al. [40] noted that T-cell behavior in the TME under ICB may be affected by mutations that affect the IFN-γ signaling pathway, particularly mutations in JAK1/2. JAK1/2 mutations have been proven to be associated with primary resistance to ICIs [69]. In addition, it has been reported that IFN-γ-induced interferon regulatory factor 8 (IRF-8) binds to its promoter and induces MDM2 overexpression [70,71]. MDM2 is a protein involved in p53 degradation and inhibition, and its amplification is often observed in HPD patients [70].

4. Monocytes

Monocytes are necessary innate immune cells which circulate in the blood and travel to tissues at sites of inflammation or infection [72]. Different monocyte subsets have completely different functions during tumor development, contributing to the activation of protumor and antitumor immune responses [73]. In addition, monocytes are the main source of macrophages and dendritic cells (DCs), which are two important components of the TME [73].
Although the expression of PD-1 in monocytes is much lower than that in T cells, monocytes can express PD-1 [74,75]. The function of PD-1 in monocytes appears to be similar to that in other types of immune cells, with PD-1 largely suppressing immune function. In fact, PD-1 is thought to negatively affect IL-12 secretion in monocytes [76,77], and anti-PD-1 treatment reverses monocyte dysfunction [78]. PD-1 expression levels in monocytes are regulated by different inflammatory stimuli; for example, IL-10 has been described as an inducer of PD-1 expression [79,80]. However, when PD-1 in monocytes is triggered by specific antibodies, it causes the monocytes to release large amounts of IL-10 [81,82]. As a well-known anti-inflammatory cytokine, IL-10 inhibits antitumor immune responses by inhibiting a variety of effector molecules and tumor cells [83].
Lu et al. [15] examined 56 metastatic gastrointestinal tract cancer patients treated with ICIs and observed that the levels of serum monocyte chemoattractant protein 1 (MCP-1) in all patients with HPD were significantly lower than those in patients without HPD. Because MCP-1 has the ability to attract monocyte aggregation, we reasonably speculate that the lack of monocytes may promote the occurrence of HPD.

5. Macrophages

Macrophages are plastic and can transform the immune environment into a protumor or antitumor environment by releasing inflammatory or inhibitory cytokines and chemokines [10]. In addition, macrophages express both PD-1 and PD-L1 in the TME and are therefore likely to be constrained by anti-PD-1/PD-L1 antibody therapy. The levels of PD-1 and PD-L1 expression in macrophages have been shown to be critical for antitumor responses in preclinical models [84].

5.1. M2 Macrophages

As shown in Figure 2, Lo Russo et al. [10] provided evidence for the reprogramming of macrophages from the M1 to M2 phenotype, leading to HPD, by studying 187 NSCLC patients who received anti-PD-1/PD-L1 treatment. The researchers observed that all HPD patients had significant numbers of CD163+CD33+PD-L1+ M2 macrophages infiltrating their tumors. This finding suggested that macrophages were involved in the progression of HPD. To prove the role of tumor-associated macrophages (TAMs) in inducing HPD, researchers also conducted in vivo experiments using multiple immunodeficient NSCLC mouse models. They treated the mice with anti-mouse PD-1 antibodies and observed increased tumor growth compared to the controls, and the macrophage abundance in the TME was similar to that observed in HPD patients. As in tumor patients, specific macrophages present in the TME may affect the development of HPD through mechanisms that do not involve the direct blockade of PD-1 in immune cells prior to the initiation of anti-PD-1 antibody therapy. In addition, other researchers have also observed that, in the treatment of NSCLC, cervical cancer, breast cancer, and colorectal cancer, anti-PD-L1 treatment may trigger the accumulation of immunosuppressive M2 macrophages characterized by a CD163+PD-L1+ phenotype at the tumor site and the accumulation of M2 macrophages may worsen the prognoses of patients treated with ICIs [85,86,87,88].
In addition, higher serum levels of ANGPT2 before ICI treatment were associated with higher levels of immunosuppressive M2 macrophages in patients treated with ICIs [89,90]. An increase in serum ANGPT2 was also observed in patients with advanced melanoma treated with anti-PD-1 antibodies [90]. Such a high level of ANGPT2 is related to an increase in the number of M2 macrophages. Therefore, ANGPT2 has been suggested as a predictor and prognostic marker in advanced melanoma patients treated with ICIs [89,90]. In addition, Wang et al. [91] demonstrated that tumor-derived exosomes induce IL-10 production in PD-1+ macrophages and block CD8+ T-cell function. Both IL-10 and ANGPT2 are contained in transcriptional signatures associated with resistance against anti-PD-1 antibodies in melanoma patients, indicating that they rapidly act on the immune regulatory mechanism and can promote immune escape and, ultimately, tumor growth [92].

5.2. FcγRIIb

Antibodies consist of the F(ab’)2 fragment, which binds antigens and the fragment crystallizable (Fc) region, which is recognized by immunoglobulin Fc receptors (FcRs) expressed by immune cells [93,94]. The interaction between the Fc domain of an antibody and its associated FcR triggers a series of immune events [95]. Most ICIs are developed by selecting antibody isotypes with low binding affinity for FcRs in order to prevent killing T cells expressing immune checkpoints [96,97,98]. In fact, both nivolumab and pembrolizumab anti-PD-1 antibodies are the IgG4 type, which is an isotype considered immunologically inert. However, they still retain the ability to bind FcγRI and FcγRIIb to activate and inhibit FcRs, respectively [96,97,98].
TAMs are capable of capturing anti-PD-1 antibodies from T-cell membranes [99]. Lo Russo et al. [10] demonstrated that nivolumab-based Fab constructs lacking Fc components did not produce HPD-like results in experimental models. The significant tumor growth observed with the full antibody was completely abolished by the use of the anti-PD-1 antibody F(ab’)2 fragment and macrophage exhaustion. In animal studies, nivolumab-associated HPD showed the infiltration of M2-like macrophages, which was suspected to be caused by the binding of Fc (nivolumab)-FCγRs. The induction of HPD by TAM reprogramming depends on the interaction of the antibody Fc with the macrophage FcR rather than the anti-PD-1 F(ab)2 fragment in the environment of tumors treated with ICIs. Since anti-PD-1 antibodies bind to the suppressive receptor FcγRIIb, researchers have hypothesized that TAMs expressing a specific subset of the suppressive receptor FcγRIIb interact with the Fc domain of anti-PD-1 antibodies to undergo functional reprogramming, thereby acquiring enhanced tumor-promoting properties and ultimately inducing HPD. Thus, anti-PD-1 antibody treatments with specific Fc sequences have the potential to promote the reprogramming of macrophages into an aggressive phenotype, leading to HPD. As shown in Figure 3, Knorr et al. [100] described how anti-PD-1 antibodies established a connection between PD-1+ macrophages and FcγRIIb+ immune cells. This interaction is speculated to induce the aggregation of several PD-1 proteins on the macrophage membrane. Close contact between different PD-1 proteins may trigger PD-1-mediated signal transduction, leading to the polarization of macrophages toward the protumor phenotype. These findings suggest that HPD can be addressed by modifying current antibody therapies aimed at disabling FcR interactions.
Although FcγRIIb may cause HPD via at least two different mechanisms, the involvement of other FcRs should not be ruled out. For example, Swisher et al. [101] reported that IgG4 could limit the response of human single-cell-derived macrophages to IFN-γ via FcγRI interactions. Activation of IgG4- mediated FcγRI shifts macrophages toward an M2-like phenotype.

6. DCs

DCs are the most effective antigen-presenting cells (APCs) that induce an effective adaptive immune response and are therefore considered to be a key factor in determining the outcomes of ICI treatment [102]. Among them, tumor infiltrating DCs (TIDCs) are a heterogeneous group of bone marrow immune cells with different mature states and functions. Some subpopulations have immunostimulatory properties and can promote antitumor immunity, while other subgroups are immunosuppressive and can facilitate tumor immune survival and tumor progression [103,104].

6.1. PD-1

PD-1 is also expressed in DCs, and the infiltration of PD-1+ TIDCs in the TME has been observed [105]. PD-1 is thought to maintain DCs in an immature phenotype and inhibit cytokine production, costimulatory molecule expression, and the antigen presentation capacity [105,106]. PD-1 expression in DCs is regulated by different stimulus signals, such as IL-10 [79]. Additionally, compensatory immune responses induced by ICB can induce the production of immunosuppressive cytokines. The blockade of PD-1+ DCs promotes increased IL-10 release from these DCs, which further enhances the regulation of PD-L1 expression in DCs, thus forming a vicious cycle that ultimately leads to enhanced immunosuppression [79]. Zhao et al. [107] demonstrated that PD-1 and PD-L1 interact simultaneously on the same cell membrane in TIDCs. In this way, PD-1 blocks PD-L1 on the same cell, thus neutralizing its ability to bind T-cell PD-1. After the administration of anti-PD-1 antibodies, PD-L1 can again be used to induce the immunosuppressive activity of T cells.

6.2. PD-L2

PD-L2 is mainly expressed in macrophages and DCs [108]. The affinity between PD-L2 and PD-1 is higher than that between PD-L1 and PD-1 [109]. The differences in clinical efficacy and toxicity between anti-PD-1 and anti-PD-L1 antibodies observed in clinical studies is likely related to the fact that anti-PD-1 antibodies can block the binding of PD-1 and PD-L2 in addition to blocking the binding of PD-1 and PD-L1. Although these ligands compete for PD-1 receptors, they are likely to initiate different responses. The role of PD-L2 has not been as thoroughly explored as that of PD-L1, but ICB may affect the interaction of PD-1 with its two ligands in different ways [110].
While PD-L1 can interchangeably bind to PD-1 or CD80 (a membrane antigen necessary for T cell activation) in T cells, PD-L2 does not bind to CD80 and induces an inhibitory TME around T-cell receptors [111]. The blockade of PD-1 may lead to the CD80/PD-L1-mediated inhibition of T cells, while PD-L1 blockade may lead to the PD-1/PD-L2-mediated inhibition of T cells [112]. The blockade of the PD-1/PD-L2 interaction leads to significant loss of inducible co-stimulator (ICOS) and CD3 expression in CD4+PD-1+ T cells, suggesting that PD-1/PD-L2 blockade disrupts the immune response in some situations [113]. Administration of soluble PD-1-Ig fusion protein inhibited bone marrow-derived DC activation and increased IL-10 secretion. The preneutralization of PD-1 with anti-PD-1 antibodies prevented this effect, suggesting that the effect is PD-1-specific and mediated by PD-L1 or PD-L2 [114].

7. Neutrophils

In a clinical research of advanced gastric cancer (AGC), Sasaki et al. [14] treated 64 patients with nivolumab and observed that C-reactive protein (CRP) and absolute neutrophil count (ANC) levels were significantly increased only in patients with HPD. They speculated that the higher CRP and ANC levels in patients with HPD might be related to the strong release of neutrophils in bone marrow and the accumulation of myeloid-derived suppressor cells (MDSCs) in tumors. In addition, abnormal inflammation observed in patients with melanoma and prostate cancer treated with anti-PD-1/PD-L1 antibodies caused by the increased Th1/Th17-dependent secretion of inflammatory cytokines, such as IL-6, IL-17, and IFN-γ, has been shown to be associated with neutrophils [115]. In addition, neutrophil exhaustion and IL-6 blockade have been shown to lead to a more effective antitumor immune response in mice [116,117,118]. Lung adenocarcinoma transgenic mice overexpressing IL-17α have stronger intrinsic resistance to anti-PD-1 antibodies, which have been demonstrated, whereas anti-IL-6 antibodies or neutrophil exhaustion lead to an effective antitumor immune response [119]. It is important to note that neutrophils generally dominate the TME in NSCLC, and their presence is closely associated with reduced infiltration of CD8+ T cells into the tumor [120]. The primary role of IL-6 and IL-17 is to promote neutrophil-mediated inflammation [121,122]. The current evidence suggests that PD-1/PD-L1 inhibition may regulate the IL-6/IL-17-neutrophil axis, resulting in abnormal inflammation, tumor immune escape, and accelerated tumor growth. Therefore, the aggregation of neutrophils in tumors after ICI treatment may lead to the generation of an immunosuppressive environment and thus promote the occurrence of HPD.
Since neutrophil infiltration may be associated with worse treatment outcomes, assessing neutrophil infiltration may be an important parameter for predicting immunotherapy outcomes [123]. One of the simplest methods is to analyze the neutrophil to lymphocyte ratio (NLR) in peripheral blood. The NLR is commonly used to assess general immune responses and systemic inflammatory states [124,125] but has also shown excellent clinical utility in ICI therapy. Mezquita et al. [126] analyzed the derived NLRs (dNLRs) in patients with NSCLC treated with chemotherapy or ICIs. A high number of circulating neutrophils was thought to be independently associated with adverse outcomes of ICI therapy but not of chemotherapy. Similar results were reported in metastatic urothelial carcinoma, and the NLR was a significant predictor of HPD [127]. Ferrara et al. [128] observed a significantly higher percentage of circulating immature low-density neutrophils in HPD patients than in non-HPD patients before ICI treatment. In addition, Gr1highLy6ClowIL-17+ neutrophil subsets were observed in mice treated with anti-PD-1 antibodies and exhibiting HPD-like tumor growth. These data suggested that neutrophils might be another indispensable factor that could promote HPD because an increased number of tumor-infiltrating neutrophils might affect the function and number of T cells, leading to a severely impaired ICI therapeutic effect.

8. MDSCs

MDSCs are a group of heterogeneous cells derived from bone marrow. They are precursors of DCs, macrophages, and granulocytes and have the ability to significantly inhibit the immune cell response [129]. PD-1 blockade negatively interacts with the innate immune system. Furthermore, MDSCs have high PD-L1 expression in the TME and weaken the efficacy of ICIs by competing with anti-PD-1 antibodies to bind T cells and secrete immunosuppressive molecules [130]. Lo Russo et al. [10] reported that MDSCs contribute to HPD, especially in the presence of M2 macrophages. In addition, IFN-γ was significantly increased after PD-1/PD-L1 blockade [131]. IFN-γ has been proven to stimulate the development of MDSCs [90] and induce the expression of indoleamine 2,3-dioxygenase, which induces the Treg cell regulation of immunosuppression in vivo [47,67]. This finding is consistent with the presence of a large number of MDSCs in the peripheral blood of HPD patients [132].

9. Natural Killer (NK) Cells

NK cells contribute to the immune attacks against tumors, but their cytotoxicity is affected by the expression level of PD-1. Anti-PD-1 antibodies not only induce the generation of powerful T cells but also restore antitumor responses in NK cells [117,133]. Solaymani-Mohammadi et al. [134] observed that a lack of PD-1 reduces the expression levels of effector molecules in NK cells, such as granzyme B and perforin, limiting the antitumor function of NK cells. Thus, complete PD-1 signaling is necessary to maintain the full function of NK cells. When PD-1 is blocked, researchers have observed the reduced production of lactic acid molecules, such as perforin and granzyme in NK cells. Therefore, NK cells may also be potential contributors to HPD.

10. Innate Lymphoid Cells (ILCs)

ILCs are a group of innate immune cells that contribute to lymph node development, tissue damage repair, antimicrobial infection, and other processes. Their dysfunction is closely related to allergic diseases, chronic infections, metabolic diseases, tumors, and other diseases [135]. ILCs are divided into several subgroups according to their characteristic surface markers, transcription factors, secreted cytokines, and immunoregulatory effects. ILC1s and ILC3s can mediate the immune response against viral, bacterial, and parasitic infections, while ILC2s are essential for maintaining tissue homeostasis [136]. The roles of these cells have been popular research topics in the field of immunology in recent years. ILCs are involved in the regulation of the innate and adaptive immune responses by secreting cytokines upon activation by different stimuli [137]. Interestingly, the immunomodulatory function of ILCs depends on secreted cytokines and the TME; thus, they play a dual role in antitumor immunity [138].
ILC3s can promote tumor progression by secreting IL-17, IL-22, and granulocyte-macrophage colony-stimulating factor (GM-CSF) [139]. Kirchberger et al. [140] reported that IL-22 produced by ILC3s reduced gastrointestinal cancer growth in colon cancer-model mice. Irshad et al. [141] provided evidence for an association between the presence of ILC3s in the TME and an increased risk of lymph node metastasis in breast cancer-model mice. Carrega et al. [142] proved that ILC3s express natural cytotoxic receptors (NCRs), which are generally considered to be expressed only in NK cells. These NCR+ ILC3s contribute to the formation of tertiary lymphoid structures (TLSs) involved with advanced tumors in patients with NSCLC. Xiong et al. [143] analyzed two patients with HPD mediated by anti-PD-1 antibodies and observed the enrichment of ILC3 marker genes. It has been reported that PD-1 is also expressed in ILC3s [144,145]. Therefore, anti-PD-1 antibodies may induce the reactivation of ILC3s and the subsequent release of cytokines, which may have protumor activity in some types of tumors [139,140]. In addition, Hepworth et al. [146] reported that ILC3s inhibited the antitumor responses of T cells by competing with T cells for IL-2, further enhancing the immunosuppression of the TME.

11. Conclusions and Future Perspectives

The advent of immunotherapy in oncology has led to unprecedented improvements in the outcomes of patients with multiple tumor types. However, evidence has indicated that immunotherapy might have a deleterious effect in some tumor patients, even promoting their disease progression. At present, many existing clinical, biological, and histopathological data that prove that a variety of immune cells modulate the effect of immunotherapy in complex ways. These findings enrich the theoretical basis of immunotherapy. Since the potential mechanisms of HPD remain uncertain, the exploration of the causative mechanisms behind HPD and the development of detection and prediction methods are urgently needed, especially in the increasingly complex clinical trials applying multiple ICIs together or an ICI in combination with other tumor treatment approaches. Further research is necessary to confirm the hypotheses that have been proposed so far and to explore other potential molecular and immunological bases of HPD to identify clear predictive factors and possibly prevent HPD. Therefore, more efforts are required in the future to inform clinical decision making for patients undergoing immunotherapy.

Author Contributions

This review was designed by Z.W. and Y.Z. This manuscript was written by Z.W. All authors have read and agreed to the published version of the manuscript.

Funding

This work was supported by grants from (1) the National Natural Science Foundation of China (No. 81571783); (2) the Capital’s Funds for Health Improvement and Research (No. 2020-2-2242) and (3) the Tsinghua Precision Medicine Foundation (No. 12020B7028).

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Data Availability Statement

Not applicable.

Acknowledgments

We thank Jiayao Liu for her help in editing the English text of a draft of this manuscript.

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Frelaut, M.; le Tourneau, C.; Borcoman, E. Hyperprogression under Immunotherapy. Int. J. Mol. Sci. 2019, 20, 2674. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  2. Shen, P.; Han, L.; Ba, X.; Qin, K.; Tu, S.H. Hyperprogressive Disease in Cancers Treated with Immune Checkpoint Inhibitors. Front. Pharmacol. 2021, 12, 678409. [Google Scholar] [CrossRef] [PubMed]
  3. Wang, X.P.; Wang, F.; Zhong, M.J.; Yarden, Y.; Fu, L.W. The biomarkers of hyperprogressive disease in PD-1/PD-L1 block-age therapy. Mol. Cancer 2020, 19, 81. [Google Scholar] [CrossRef]
  4. Wolchok, J.D.; Hoos, A.; O’Day, S.; Weber, J.S.; Hamid, O.; Lebbé, C.; Maio, M.; Binder, M.; Bohnsack, O.; Nichol, G.; et al. Guidelines for the Evaluation of Immune Therapy Activity in Solid Tumors: Immune-Related Response Criteria. Clin. Cancer Res. 2009, 15, 7412–7420. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  5. Lin, M.; Vanneste, B.G.L.; Yu, Q.; Chen, Z.; Peng, J.; Cai, X. Hyperprogression under immunotherapy: A new form of immunotherapy response?—A narrative literature review. Transl. Lung Cancer Res. 2021, 10, 3276–3291. [Google Scholar] [CrossRef] [PubMed]
  6. Champiat, S.; Dercle, L.; Ammari, S.; Massard, C.; Hollebecque, A.; Postel-Vinay, S.; Chaput, N.; Eggermont, A.; Marabelle, A.; Soria, J.-C.; et al. Hyperprogressive Disease Is a New Pattern of Progression in Cancer Patients Treated by Anti-PD-1/PD-L1. Clin. Cancer Res. 2017, 23, 1920–1928. [Google Scholar] [CrossRef] [Green Version]
  7. Saada-Bouzid, E.; Defaucheux, C.; Karabajakian, A.; Coloma, V.P.; Servois, V.; Paoletti, X.; Even, C.; Fayette, J.; Guigay, J.; Loirat, D.; et al. Hyperprogression during anti-PD-1/PD-L1 therapy in patients with recurrent and/or metastatic head and neck squamous cell carcinoma. Ann. Oncol. 2017, 28, 1605–1611. [Google Scholar] [CrossRef]
  8. Ferrara, R.; Mezquita, L.; Texier, M.; Lahmar, J.; Audigier-Valette, C.; Tessonnier, L.; Mazieres, J.; Zalcman, G.; Brosseau, S.; Le Moulec, S.; et al. Hyperprogressive Disease in Patients with Advanced Non-Small Cell Lung Cancer Treated with PD-1/PD-L1 Inhibitors or with Single-Agent Chemotherapy. JAMA Oncol. 2018, 4, 1543–1552. [Google Scholar] [CrossRef]
  9. Kanazu, M.; Edahiro, R.; Krebe, H.; Nishida, K.; Ishijima, M.; Uenami, T.; Akazawa, Y.; Yano, Y.; Yamaguchi, T.; Mori, M. Hyperprogressive disease in patients with non-small cell lung cancer treated with nivolumab: A case series. Thorac. Cancer 2018, 9, 1782–1787. [Google Scholar] [CrossRef] [Green Version]
  10. Russo, G.L.; Moro, M.; Sommariva, M.; Cancila, V.; Boeri, M.; Centonze, G.; Ferro, S.; Ganzinelli, M.; Gasparini, P.; Huber, V.; et al. Antibody-Fc/FcR Interac-tion on Macrophages as a Mechanism for Hyperprogressive Disease in Non-small Cell Lung Cancer Subsequent to PD-1/PD-L1 Blockade. Clin. Cancer Res. 2019, 25, 989–999. [Google Scholar] [CrossRef] [Green Version]
  11. Kim, C.G.; Kim, K.H.; Pyo, K.H.; Xin, C.F.; Hong, M.H.; Ahn, B.C.; Kim, Y.; Choi, S.J.; Yoon, H.I.; Lee, J.G.; et al. Hyperprogressive disease during PD-1/PD-L1 blockade in patients with non-small-cell lung cancer. Ann. Oncol. 2019, 30, 1104–1113. [Google Scholar] [CrossRef] [PubMed]
  12. Aoki, M.; Shoji, H.; Nagashima, K.; Imazeki, H.; Miyamoto, T.; Hirano, H.; Honma, Y.; Iwasa, S.; Okita, N.; Takashima, A.; et al. Hyperprogressive disease during nivolumab or irinotecan treatment in patients with advanced gastric cancer. ESMO Open 2019, 4, e000488. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  13. Kanjanapan, Y.; Day, D.; Wang, L.; Al-Sawaihey, H.; Abbas, E.; Namini, A.; Siu, L.L.; Hansen, A.; Razak, A.A.; Spreafico, A.; et al. Hyperprogressive disease in early-phase immunotherapy trials: Clinical predictors and association with im-mune-related toxicities. Cancer 2019, 125, 1341–1349. [Google Scholar] [CrossRef] [PubMed]
  14. Sasaki, A.; Nakamura, Y.; Mishima, S.; Kawazoe, A.; Kuboki, Y.; Bando, H.; Kojima, T.; Doi, T.; Ohtsu, A.; Yoshino, T.; et al. Predictive factors for hyperprogressive disease during nivolumab as anti-PD1 treatment in patients with advanced gastric cancer. Gastric Cancer 2019, 22, 793–802. [Google Scholar] [CrossRef] [Green Version]
  15. Lu, Z.; Zou, J.; Hu, Y.; Li, S.; Zhou, T.; Gong, J.; Li, J.; Zhang, X.; Zhou, J.; Lu, M.; et al. Serological Markers Associated with Response to Immune Checkpoint Blockade in Metastatic Gastrointestinal Tract Cancer. JAMA Netw. Open 2019, 2, e197621. [Google Scholar] [CrossRef] [Green Version]
  16. Matos, I.; Martin-Liberal, J.; García-Ruiz, A.; Hierro, C.; de Olza, M.O.; Viaplana, C.; Azaro, A.; Vieito, M.; Braña, I.; Mur, G.; et al. Capturing Hyperprogressive Disease with Immune-Checkpoint Inhibitors Using RECIST 1.1 Criteria. Clin. Cancer Res. 2020, 26, 1846–1855. [Google Scholar] [CrossRef] [Green Version]
  17. Castello, A.; Rossi, S.; Mazziotti, E.; Toschi, L.; Lopci, E. Hyperprogressive Disease in Patients with Non–Small Cell Lung Cancer Treated with Checkpoint Inhibitors: The Role of 18F-FDG PET/CT. J. Nucl. Med. 2020, 61, 821–826. [Google Scholar] [CrossRef]
  18. Hwang, I.; Park, I.; Yoon, S.-k.; Lee, J.L. Hyperprogressive Disease in Patients with Urothelial Carcinoma or Renal Cell Carcinoma Treated with PD-1/PD-L1 Inhibitors. Clin. Genitourin. Cancer 2020, 18, E122–E133. [Google Scholar] [CrossRef]
  19. Vaidya, P.; Bera, K.; Patil, P.D.; Gupta, A.; Jain, P.; Alilou, M.; Khorrami, M.; Velcheti, V.; Madabhushi, A. Novel, non-invasive imaging approach to identify patients with advanced non-small cell lung cancer at risk of hyperprogressive disease with immune checkpoint blockade. J. Immunother. Cancer 2020, 8, e001343. [Google Scholar] [CrossRef]
  20. Zhang, L.; Wu, L.; Chen, Q.; Zhang, B.; Liu, J.; Liu, S.; Mo, X.; Li, M.; Chen, Z.; Chen, L.; et al. Predicting hyperprogressive disease in patients with advanced hepatocellular carcinoma treated with anti-programmed cell death 1 therapy. eClinicalMedicine 2021, 31, 100673. [Google Scholar] [CrossRef]
  21. Kim, C.G.; Kim, C.; Yoon, S.E.; Kim, K.H.; Choi, S.J.; Kang, B.; Kim, H.R.; Park, S.-H.; Shin, E.-C.; Kim, Y.-Y.; et al. Hyperprogressive disease during PD-1 blockade in patients with advanced hepatocellular carcinoma. J. Hepatol. 2021, 74, 350–359. [Google Scholar] [CrossRef] [PubMed]
  22. Chen, S.; Gou, M.; Yan, H.; Fan, M.; Pan, Y.; Fan, R.; Qian, N.; Dai, G. Hyperprogressive Disease Caused by PD-1 Inhibitors for the Treatment of Pan-Cancer. Dis. Markers 2021, 2021, 6639366. [Google Scholar] [CrossRef] [PubMed]
  23. Xiao, L.-S.; Li, Q.-M.; Hu, C.-Y.; Cui, H.; Hong, C.; Huang, C.-Y.; Li, R.-N.; Dong, Z.-Y.; Zhu, H.-B.; Liu, L. Lung metastasis and lymph node metastasis are risk factors for hyperprogressive disease in primary liver cancer patients treated with immune checkpoint inhibitors. Ann. Palliat. Med. 2021, 10, 11244–11254. [Google Scholar] [CrossRef] [PubMed]
  24. Miyama, Y.; Morikawa, T.; Miyakawa, J.; Koyama, Y.; Kawai, T.; Kume, H.; Ushiku, T. Squamous differentiation is a potential biomarker predicting tumor progression in patients treated with pembrolizumab for urothelial carcinoma. Pathol.-Res. Pract. 2021, 219, 153364. [Google Scholar] [CrossRef] [PubMed]
  25. Maesaka, K.; Sakamori, R.; Yamada, R.; Tahata, Y.; Imai, Y.; Ohkawa, K.; Miyazaki, M.; Mita, E.; Ito, T.; Hagiwara, H.; et al. Hyperprogressive disease in patients with unresectable hepatocellular carcinoma receiving atezolizumab plus bevacizumab therapy. Hepatol. Res. 2022, 52, 298–307. [Google Scholar] [CrossRef]
  26. Tay, C.; Qian, Y.; Sakaguchi, S. Hyper-Progressive Disease: The Potential Role and Consequences of T-Regulatory Cells Foiling Anti-PD-1 Cancer Immunotherapy. Cancers 2021, 13, 48. [Google Scholar] [CrossRef]
  27. Pardoll, D.M. The blockade of immune checkpoints in cancer immunotherapy. Nat. Rev. Cancer 2012, 12, 252–264. [Google Scholar] [CrossRef] [Green Version]
  28. Wherry, E.J.; Kurachi, M. Molecular and cellular insights into T cell exhaustion. Nat. Rev. Immunol. 2015, 15, 486–499. [Google Scholar] [CrossRef]
  29. Franco, F.; Jaccard, A.; Romero, P.; Yu, Y.-R.; Ho, P.-C. Metabolic and epigenetic regulation of T-cell exhaustion. Nat. Metab. 2020, 2, 1001–1012. [Google Scholar] [CrossRef]
  30. Crespo, J.; Sun, H.Y.; Welling, T.H.; Tian, Z.G.; Zou, W.P. T cell anergy, exhaustion, senescence, and stemness in the tumor microenvironment. Curr. Opin. Immunol. 2013, 25, 214–221. [Google Scholar] [CrossRef] [Green Version]
  31. Matsuzaki, J.; Gnjatic, S.; Mhawech-Fauceglia, P.; Beck, A.; Miller, A.; Tsuji, T.; Eppolito, C.; Qian, F.; Lele, S.; Shrikant, P.; et al. Tumor-infiltrating NY-ESO-1–specific CD8 + T cells are negatively regulated by LAG-3 and PD-1 in human ovarian cancer. Proc. Natl. Acad. Sci. USA 2010, 107, 7875–7880. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  32. Baitsch, L.; Baumgaertner, P.; Devêvre, E.; Raghav, S.K.; Legat, A.; Barba, L.; Wieckowski, S.; Bouzourene, H.; Deplancke, B.; Romero, P.; et al. Exhaustion of tumor-specific CD8+ T cells in metastases from melanoma patients. J. Clin. Investig. 2011, 121, 2350–2360. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  33. Thommen, D.S.; Schreiner, J.; Müller, P.; Herzig, P.; Roller, A.; Belousov, A.; Umana, P.; Pisa, P.; Klein, C.G.; Bacac, M.; et al. Progression of Lung Cancer Is Associated with Increased Dysfunction of T Cells Defined by Coexpression of Multiple Inhibitory Receptors. Cancer Immunol. Res. 2015, 3, 1344–1355. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  34. Lu, X.; Yang, L.; Yao, D.X.; Wu, X.; Li, J.P.; Liu, X.S.; Deng, L.J.; Huang, C.T.; Wang, Y.; Li, D.; et al. Tumor anti-gen-specific CD8+ T cells are negatively regulated by PD-1 and Tim-3 in human gastric cancer. Cell. Immunol. 2017, 313, 43–51. [Google Scholar] [CrossRef]
  35. Odorizzi, P.M.; Pauken, K.E.; Paley, M.A.; Sharpe, A.H.; Wherry, E.J. Genetic absence of PD-1 promotes accumulation of terminally differentiated exhausted CD8+ T cells. J. Exp. Med. 2015, 212, 1125–1137. [Google Scholar] [CrossRef] [PubMed]
  36. Wartewig, T.; Kurgyis, Z.; Keppler, S.J.; Pechloff, K.; Hameister, E.; Öllinger, R.; Maresch, R.; Buch, T.; Steiger, K.; Winter, C.; et al. PD-1 is a haploinsufficient suppressor of T cell lymphomagenesis. Nature 2017, 552, 121–125. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  37. Ratner, L.; Waldmann, T.A.; Janakiram, M.; Brammer, J.E. Rapid Progression of Adult T-Cell Leukemia–Lymphoma after PD-1 Inhibitor Therapy. N. Engl. J. Med. 2018, 378, 1947–1948. [Google Scholar] [CrossRef]
  38. Stein, R.G.; Ebert, S.; Schlahsa, L.; Scholz, C.J.; Braun, M.; Hauck, P.; Horn, E.; Monoranu, C.-M.; Thiemann, V.J.; Wustrow, M.P.; et al. Cognate Nonlytic Interactions between CD8+ T Cells and Breast Cancer Cells Induce Cancer Stem Cell–like Properties. Cancer Res. 2019, 79, 1507–1519. [Google Scholar] [CrossRef] [Green Version]
  39. Gil Del Alcazar, C.R.; Huh, S.J.; Ekram, M.B.; Trinh, A.; Liu, L.L.; Beca, F.; Zi, X.; Kwak, M.; Bergholtz, H.; Su, Y.; et al. Immune Escape in Breast Cancer During In Situ to Invasive Carcinoma Transition. Cancer Discov. 2017, 7, 1098–1115. [Google Scholar] [CrossRef] [Green Version]
  40. Cimino-Mathews, A.; Foote, J.B.; Emens, L.A. Immune Targeting in Breast Cancer. Oncology 2015, 29, 375–385. [Google Scholar]
  41. Al-Hajj, M.; Wicha, M.S.; Benito-Hernandez, A.; Morrison, S.J.; Clarke, M.F. Prospective identification of tumorigenic breast cancer cells. Proc. Natl. Acad. Sci. USA 2003, 100, 3983–3988. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  42. Reim, F.; Dombrowski, Y.; Ritter, C.; Buttmann, M.; Hausler, S.; Ossadnik, M.; Krockenberger, M.; Beier, D.; Beier, C.P.; Dietl, J.; et al. Immunoselection of Breast and Ovarian Cancer Cells with Trastuzumab and Natural Killer Cells: Selective Escape of CD44(high)/CD24(low)/HER2(low) Breast Cancer Stem Cells. Cancer Res. 2009, 69, 8058–8066. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  43. Jiang, W.Q.; He, Y.J.; He, W.G.; Wu, G.S.; Zhou, X.L.; Sheng, Q.S.; Zhong, W.X.; Lu, Y.M.; Ding, Y.F.; Lu, Q.; et al. Exhausted CD8+T Cells in the Tumor Immune Microenvironment: New Pathways to Therapy. Front. Immunol. 2021, 11, 622509. [Google Scholar] [CrossRef] [PubMed]
  44. Huang, R.-Y.; Francois, A.; McGray, A.R.; Miliotto, A.; Odunsi, K. Compensatory upregulation of PD-1, LAG-3, and CTLA-4 limits the efficacy of single-agent checkpoint blockade in metastatic ovarian cancer. OncoImmunology 2016, 6, e1249561. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  45. YKawakami; Ohta, S.; Sayem, M.A.; Tsukamoto, N.; Yaguchi, T. Immune-resistant mechanisms in cancer immunothera-py. Int. J. Clin. Oncol. 2020, 25, 810–817. [Google Scholar] [CrossRef]
  46. Shi, H.B.; Lan, J.; Yang, J.Q. Mechanisms of Resistance to Checkpoint Blockade Therapy. Adv Exp Med Biol. 2020, 1248, 83–117. [Google Scholar]
  47. Camelliti, S.; Le Noci, V.; Bianchi, F.; Moscheni, C.; Arnaboldi, F.; Gagliano, N.; Balsari, A.; Garassino, M.C.; Tagliabue, E.; Sfondrini, L.; et al. Mechanisms of hyperprogressive disease after immune checkpoint inhibitor therapy: What we (don’t) know. J. Exp. Clin. Cancer Res. 2020, 39, 236. [Google Scholar] [CrossRef]
  48. Koyama, S.; Akbay, E.A.; Li, Y.Y.; Aref, A.R.; Skoulidis, F.; Herter-Sprie, G.S.; Buczkowski, K.A.; Liu, Y.; Awad, M.M.; Denning, W.L.; et al. STK11/LKB1 Deficiency Promotes Neutrophil Recruitment and Proinflammatory Cytokine Production to Suppress T-cell Activity in the Lung Tumor Microenvironment. Cancer Res. 2016, 76, 999–1008. [Google Scholar] [CrossRef] [Green Version]
  49. Sun, Z.; Fourcade, J.; Pagliano, O.; Chauvin, J.-M.; Sander, C.; Kirkwood, J.M.; Zarour, H.M. IL10 and PD-1 Cooperate to Limit the Activity of Tumor-Specific CD8+ T Cells. Cancer Res. 2015, 75, 1635–1644. [Google Scholar] [CrossRef] [Green Version]
  50. Richardson, J.R.; Schollhorn, A.; Gouttefangeas, C.; Schuhmacher, J. CD4+T Cells: Multitasking Cells in the Duty of Cancer Immunotherapy. Cancers 2021, 13, 596. [Google Scholar] [CrossRef]
  51. Togashi, Y.; Shitara, K.; Nishikawa, H. Regulatory T cells in cancer immunosuppression—Implications for anticancer therapy. Nat. Rev. Clin. Oncol. 2019, 16, 356–371. [Google Scholar] [CrossRef] [PubMed]
  52. Kamada, T.; Togashi, Y.; Tay, C.; Ha, D.; Sasaki, A.; Nakamura, Y.; Sato, E.; Fukuoka, S.; Tada, Y.; Tanaka, A.; et al. PD-1+ regulatory T cells amplified by PD-1 blockade promote hyperprogression of cancer. Proc. Natl. Acad. Sci. USA 2019, 116, 9999–10008. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  53. Cai, J.J.; Wang, D.S.; Zhang, G.Y.; Guo, X.L. The Role Of PD-1/PD-L1 Axis In Treg Development And Function: Implica-tions For Cancer Immunotherapy. Oncotargets Ther. 2019, 12, 8437–8445. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  54. Rauch, D.A.; Conlon, K.C.; Janakiram, M.; Brammer, J.E.; Harding, J.C.; Ye, B.H.; Zang, X.; Ren, X.; Olson, S.; Cheng, X.; et al. Rapid progression of adult T-cell leukemia/lymphoma as tumor-infiltrating Tregs after PD-1 blockade. Blood 2019, 134, 1406–1414. [Google Scholar] [CrossRef]
  55. Simpson, T.R.; Li, F.; Montalvo-Ortiz, W.; Sepulveda, M.A.; Bergerhoff, K.; Arce, F.; Roddie, C.; Henry, J.Y.; Yagita, H.; Wolchok, J.D.; et al. Fc-dependent depletion of tumor-infiltrating regulatory T cells co-defines the efficacy of anti–CTLA-4 therapy against melanoma. J. Exp. Med. 2013, 210, 1695–1710. [Google Scholar] [CrossRef]
  56. Ji, Z.; Peng, Z.; Gong, J.; Zhang, X.; Li, J.; Lu, M.; Lu, Z.; Shen, L. Hyperprogression after immunotherapy in patients with malignant tumors of digestive system. BMC Cancer 2019, 19, 1–9. [Google Scholar] [CrossRef] [Green Version]
  57. Champiat, S.; Ferrara, R.; Massard, C.; Besse, B.; Marabelle, A.; Soria, J.-C.; Ferté, C. Hyperprogressive disease: Recognizing a novel pattern to improve patient management. Nat. Rev. Clin. Oncol. 2018, 15, 748–762. [Google Scholar] [CrossRef]
  58. Lee, G.H.; Lee, W.-W. Unusual CD4+CD28T Cells and Their Pathogenic Role in Chronic Inflammatory Disorders. Immune Netw. 2016, 16, 322–329. [Google Scholar] [CrossRef] [Green Version]
  59. Maly, K.; Schirmer, M. The Story of CD4+ CD28 T Cells Revisited: Solved or Still Ongoing? J. Immunol. Res. 2015, 2015, 348746. [Google Scholar]
  60. Arasanz, H.; Zuazo, M.; Bocanegra, A.; Gato, M.; Martinez-Aguillo, M.; Morilla, I.; Fernandez, G.; Hernandez, B.; Lopez, P.; Alberdi, N.; et al. Early Detection of Hyperprogressive Dis-ease in Non-Small Cell Lung Cancer by Monitoring of Systemic T Cell Dynamics. Cancers 2020, 12, 344. [Google Scholar] [CrossRef] [Green Version]
  61. Zappasodi, R.; Budhu, S.; Hellmann, M.D.; Postow, M.A.; Senbabaoglu, Y.; Manne, S.; Gasmi, B.; Liu, C.L.; Zhong, H.; Li, Y.Y.; et al. Non-conventional Inhibi-tory CD4(+)Foxp3(-)PD-1(hi) T Cells as a Biomarker of Immune Checkpoint Blockade Activity. Cancer Cell 2018, 33, 1017–1032.e7. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  62. Miggelbrink, A.M.; Jackson, J.D.; Lorrey, S.J.; Srinivasan, E.S.; Waibl-Polania, J.; Wilkinson, D.S.; Fecci, P.E. CD4 T-Cell Exhaustion: Does It Exist and What Are Its Roles in Cancer? Clin. Cancer Res. 2021, 27, 5742–5752. [Google Scholar] [CrossRef] [PubMed]
  63. Martini, M.; Testi, M.G.; Pasetto, M.; Picchio, M.C.; Innamorati, G.; Mazzocco, M.; Ugel, S.; Cingarlini, S.; Bronte, V.; Zanovello, P.; et al. IFN-gamma-mediated upmodulation of MHC class I expression activates tumor-specific immune response in a mouse model of prostate cancer. Vaccine 2010, 28, 3548–3557. [Google Scholar] [CrossRef] [PubMed]
  64. Zimmerman, M.; Yang, D.F.; Hu, X.L.; Liu, F.Y.; Singh, N.; Browning, D.; Ganapathy, V.; Chandler, P.; Choubey, D.; Abrams, S.I.; et al. IFN-gamma Upregulates Survivin and Ifi202 Expression to Induce Survival and Proliferation of Tumor-Specific T Cells. PLoS ONE 2010, 5. [Google Scholar] [CrossRef]
  65. Xiao, M.J.; Wang, C.H.; Zhang, J.H.; Li, Z.G.; Zhao, X.Q.; Qin, Z.H. IFN gamma Promotes Papilloma Development by Up-regulating Th17-Associated Inflammation. Cancer Res. 2009, 69, 2010–2017. [Google Scholar] [CrossRef] [Green Version]
  66. O’Garra, A.; Barrat, F.J.; Castro, G.; Vicari, A.; Hawrylowicz, C. Strategies for use of IL-10 or its antagonists in human disease. Immunol. Rev. 2008, 223, 114–131. [Google Scholar] [CrossRef]
  67. Sakai, S.; Kauffman, K.D.; Sallin, M.A.; Sharpe, A.H.; Young, H.A.; Ganusov, V.V.; Barber, D.L. CD4 T Cell-Derived IFN-gamma Plays a Minimal Role in Control of Pulmonary Mycobacterium tuberculosis Infection and Must Be Actively Re-pressed by PD-1 to Prevent Lethal Disease. PLoS Pathog. 2016, 12, e1005667. [Google Scholar] [CrossRef] [Green Version]
  68. Rosenkranz, D.; Weyer, S.; Tolosa, E.; Gaenslen, A.; Berg, D.; Leyhe, T.; Gasser, T.; Stoltze, L. Higher frequency of regulatory T suppressive activity cells in the elderly and increased in neurodegeneration. J. Neuroimmunol. 2007, 188, 117–127. [Google Scholar] [CrossRef]
  69. Shin, D.S.; Zaretsky, J.M.; Escuin-Ordinas, H.; Garcia-Diaz, A.; Hu-Lieskovan, S.; Kalbasi, A.; Grasso, C.S.; Hugo, W.; Sandoval, S.; Torrejon, D.Y.; et al. Primary Resistance to PD-1 Blockade Mediated by JAK1/2 Mutations. Cancer Discov. 2017, 7, 188–201. [Google Scholar] [CrossRef] [Green Version]
  70. Kato, S.; Goodman, A.; Walavalkar, V.; Barkauskas, D.A.; Sharabi, A.; Kurzrock, R. Hyperprogressors after Immunothera-py: Analysis of Genomic Alterations Associated with Accelerated Growth Rate. Clin. Cancer Res. 2017, 23, 4242–4250. [Google Scholar] [CrossRef] [Green Version]
  71. Zhou, J.X.; Lee, C.H.; Qi, C.F.; Wang, H.; Naghashfar, Z.; Abbasi, S.; Morse, H.C. IFN Regulatory Factor 8 Regulates MDM2 in Germinal Center B Cells. J. Immunol. 2009, 183, 3188–3194. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  72. Kratofil, R.M.; Kubes, P.; Deniset, J.F. Monocyte Conversion During Inflammation and Injury. Arterioscler. Thromb. Vasc. Biol. 2017, 37, 35–42. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  73. Olingy, C.E.; Dinh, H.Q.; Hedrick, C.C. Monocyte heterogeneity and functions in cancer. J. Leukoc. Biol. 2019, 106, 309–322. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  74. Zasada, M.; Lenart, M.; Rutkowska-Zapała, M.; Stec, M.; Durlak, W.; Grudzień, A.; Krzeczkowska, A.; Mól, N.; Pilch, M.; Siedlar, M.; et al. Analysis of PD-1 expression in the monocyte subsets from non-septic and septic preterm neonates. PLoS ONE 2017, 12, e0186819. [Google Scholar] [CrossRef] [Green Version]
  75. da Mota, N.V.F.; Brunialti, M.K.C.; Santos, S.S.; Machado, F.R.; Assuncao, M.; Azevedo, L.C.P.; Salomao, R. Immuno-phenotyping of monocytes during human sepsis shows impairment in antigen presentation: A shift toward nonclassical differentiation and upregulation of fc gamma ri-receptor. Shock 2018, 50, 293–300. [Google Scholar] [CrossRef]
  76. Ma, C.J.; Ni, L.; Zhang, Y.; Zhang, C.L.; Wu, X.Y.; Atia, A.N.; Thayer, P.; Moorman, J.P.; Yao, Z.Q. PD-1 negatively regulates interleukin-12 expression by limiting STAT-1 phosphorylation in monocytes/macrophages duringchronic hepatitis C virus in-fection. Immunology 2011, 132, 421–431. [Google Scholar] [CrossRef]
  77. Zhang, Y.; Ma, C.J.; Ni, L.; Zhang, C.L.; Wu, X.Y.; Kumaraguru, U.; Li, C.F.; Moorman, J.P.; Yao, Z.Q. Cross-Talk between Programmed Death-1 and Suppressor of Cytokine Signaling-1 in Inhibition of IL-12 Production by Monocytes/Macrophages in Hepatitis C Virus Infection. J. Immunol. 2011, 186, 3093–3103. [Google Scholar] [CrossRef] [Green Version]
  78. Zhang, Y.; Zhou, Y.; Lou, J.; Li, J.; Bo, L.; Zhu, K.; Wan, X.; Deng, X.; Cai, Z. PD-L1 blockade improves survival in experimental sepsis by inhibiting lymphocyte apoptosis and reversing monocyte dysfunction. Crit. Care 2010, 14, R220. [Google Scholar] [CrossRef] [Green Version]
  79. Lamichhane, P.; Karyampudi, L.; Shreeder, B.; Krempski, J.; Bahr, D.; Daum, J.; Kalli, K.R.; Goode, E.L.; Block, M.S.; Cannon, M.J.; et al. IL10 Release upon PD-1 Blockade Sustains Immunosuppression in Ovarian Cancer. Cancer Res. 2017, 77, 6667–6678. [Google Scholar] [CrossRef] [Green Version]
  80. Ka, M.B.; Gondois-Rey, F.; Capo, C.; Textoris, J.; Million, M.; Raoult, D.; Olive, D.; Mege, J.L. Imbalance of Circulating Mon-ocyte Subsets and PD-1 Dysregulation in Q Fever Endocarditis: The Role of IL-10 in PD-1 Modulation. PLoS ONE 2014, 9, e107533. [Google Scholar] [CrossRef]
  81. Said, E.A.; Dupuy, F.P.; Trautmann, L.; Zhang, Y.W.; Shi, Y.; El-Far, M.; Hill, B.J.; Noto, A.; Ancuta, P.; Peretz, Y.; et al. Pro-grammed death-1-induced interleukin-10 production by monocytes impairs CD4(+) T cell activation during HIV infection. Nat. Med. 2010, 16, 452–459. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  82. Xia, Q.; Wei, L.; Zhang, Y.; Sheng, J.; Wu, W.; Zhang, Y. Immune Checkpoint Receptors Tim-3 and PD-1 Regulate Monocyte and T Lymphocyte Function in Septic Patients. Mediat. Inflamm. 2018, 2018, 1632902. [Google Scholar] [CrossRef] [PubMed]
  83. Ouyang, W.; O’Garra, A. IL-10 Family Cytokines IL-10 and IL-22: From Basic Science to Clinical Translation. Immunity 2019, 50, 871–891. [Google Scholar] [CrossRef] [PubMed]
  84. Gordon, S.R.; Maute, R.L.; Dulken, B.W.; Hutter, G.; George, B.M.; McCracken, M.N.; Gupta, R.; Tsai, J.M.; Sinha, R.; Corey, D.; et al. PD-1 expression by tumour-associated macrophages inhibits phagocytosis and tumour immunity. Nature 2017, 545, 495–499. [Google Scholar] [CrossRef]
  85. Lavin, Y.; Kobayashi, S.; Leader, A.; Amir, E.D.; Elefant, N.; Bigenwald, C.; Remark, R.; Sweeney, R.; Becker, C.D.; Lev-ine, J.H.; et al. Innate Immune Landscape in Early Lung Adenocarcinoma by Paired Single-Cell Analyses. Cell 2017, 169, 750–765. [Google Scholar] [CrossRef] [Green Version]
  86. Korehisa, S.; Oki, E.; Iimori, M.; Nakaji, Y.; Shimokawa, M.; Saeki, H.; Okano, S.; Oda, Y.; Maehara, Y. Clinical significance of programmed cell death-ligand 1 expression and the immune microenvironment at the invasive front of colorectal cancers with high microsatellite instability. Int. J. Cancer 2017, 142, 822–832. [Google Scholar] [CrossRef] [Green Version]
  87. Adams, T.A.; Vail, P.J.; Ruiz, A.; Mollaee, M.; McCue, P.A.; Knudsen, E.S.; Witkiewicz, A.K. Composite analysis of immu-nological and metabolic markers defines novel subtypes of triple negative breast cancer. Mod. Pathol. 2018, 31, 288–298. [Google Scholar] [CrossRef]
  88. Harada, K.; Dong, X.; Estrella, J.S.; Correa, A.M.; Xu, Y.; Hofstetter, W.L.; Sudo, K.; Onodera, H.; Suzuki, K.; Suzuki, A.; et al. Tumor-associated macrophage infiltration is highly associated with PD-L1 expression in gastric adenocarcinoma. Gastric Cancer 2017, 21, 31–40. [Google Scholar] [CrossRef]
  89. Scholz, A.; Lang, V.; Henschler, R.; Czabanka, M.; Vajkoczy, P.; Chavakis, E.; Drynski, J.; Harter, P.N.; Mittelbronn, M.; Dumont, D.J.; et al. Angiopoietin-2 promotes myeloid cell infiltration in a beta(2)-integrin-dependent manner. Blood 2011, 118, 5050–5059. [Google Scholar] [CrossRef]
  90. Wu, X.Q.; Giobbie-Hurder, A.; Liao, X.Y.; Connelly, C.; Connolly, E.M.; Li, J.J.; Manos, M.P.; Lawrence, D.; McDermott, D.; Severgnini, M.; et al. Angiopoietin-2 as a Bi-omarker and Target for Immune Checkpoint Therapy. Cancer Immunol. Res. 2017, 5, 17–28. [Google Scholar] [CrossRef] [Green Version]
  91. Wang, F.; Li, B.; Wei, Y.; Zhao, Y.; Wang, L.; Zhang, P.; Yang, J.; He, W.; Chen, H.; Jiao, Z.; et al. Tumor-derived exosomes induce PD1+ macrophage population in human gastric cancer that promotes disease progression. Oncogenesis 2018, 7, 1–11. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  92. Hugo, W.; Zaretsky, J.M.; Sun, L.; Song, C.; Moreno, B.H.; Hu-Lieskovan, S.; Berent-Maoz, B.; Pang, J.; Chmielowski, B.; Cherry, G.; et al. Genomic and Transcriptomic Features of Response to Anti-PD-1 Therapy in Metastatic Melanoma. Cell 2016, 165, 35–44. [Google Scholar] [CrossRef] [Green Version]
  93. Kang, T.H.; Jung, S.T. Boosting therapeutic potency of antibodies by taming Fc domain functions. Exp. Mol. Med. 2019, 51, 1–9. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  94. Chiu, M.L.; Goulet, D.R.; Teplyakov, A.; Gilliland, G.L. Antibody Structure and Function: The Basis for Engineering Therapeutics. Antibodies 2019, 8, 55. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  95. Vidarsson, G.; Dekkers, G.; Rispens, T. IgG Subclasses and Allotypes: From Structure to Effector Functions. Front. Immunol. 2014, 5, 520. [Google Scholar] [CrossRef] [Green Version]
  96. Kretschmer, A.; Schwanbeck, R.; Valerius, T.; Rosner, T. Antibody Isotypes for Tumor Immunotherapy. Transfus. Med. Hemotherapy 2017, 44, 320–326. [Google Scholar] [CrossRef] [Green Version]
  97. Chen, X.; Song, X.M.; Li, K.; Zhang, T. Fc gamma R-Binding Is an Important Functional Attribute for Immune Checkpoint Antibodies in Cancer Immunotherapy. Front. Immunol. 2019, 10, 292. [Google Scholar] [CrossRef]
  98. Yu, J.; Song, Y.; Tian, W. How to select IgG subclasses in developing anti-tumor therapeutic antibodies. J. Hematol. Oncol. 2020, 13, 45. [Google Scholar] [CrossRef]
  99. Arlauckas, S.P.; Garris, C.S.; Kohler, R.H.; Kitaoka, M.; Cuccarese, M.F.; Yang, K.S.; Miller, M.A.; Carlson, J.C.; Freeman, G.J.; Anthony, R.M.; et al. In vivo imaging reveals a tumor-associated macrophage–mediated resistance pathway in anti–PD-1 therapy. Sci. Transl. Med. 2017, 9, eaal3604. [Google Scholar] [CrossRef] [Green Version]
  100. Knorr, D.A.; Ravetch, J.V. Immunotherapy and Hyperprogression: Unwanted Outcomes, Unclear Mechanism. Clin. Cancer Res. 2019, 25, 904–906. [Google Scholar] [CrossRef] [Green Version]
  101. Swisher, J.F.A.; Haddad, D.A.; McGrath, A.G.; Boekhoudt, G.H.; Feldman, G.M. IgG4 can induce an M2-like phenotype in human monocyte-derived macrophages through Fc gamma RI. Mabs 2014, 6, 1377–1384. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  102. Versteven, M.; Van den Bergh, J.M.J.; Marcq, E.; Smits, E.L.J.; Van Tendeloo, V.F.I.; Hobo, W.; Lion, E. Dendritic Cells and Programmed Death-1 Blockade: A Joint Venture to Combat Cancer. Front. Immunol. 2018, 9, 394. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  103. Laoui, D.; Keirsse, J.; Morias, Y.; van Overmeire, E.; Geeraerts, X.; Elkrim, Y.; Kiss, M.; Bolli, E.; Lahmar, Q.; Sichien, D.; et al. The tumour microenviron-ment harbours ontogenically distinct dendritic cell populations with opposing effects on tumour immunity. Nat. Commun. 2016, 7, 13720. [Google Scholar]
  104. Keirsse, J.; Van Damme, H.; Van Ginderachter, J.A.; Laoui, D. Exploiting tumor-associated dendritic cell heterogeneity for novel cancer therapies. J. Leukoc. Biol. 2017, 102, 317–324. [Google Scholar] [CrossRef]
  105. Krempski, J.; Karyampudi, L.; Behrens, M.D.; Erskine, C.L.; Hartmann, L.; Dong, H.; Goode, E.L.; Kalli, K.R.; Knutson, K.L. Tumor-Infiltrating Programmed Death Receptor-1+ Dendritic Cells Mediate Immune Suppression in Ovarian Cancer. J. Immunol. 2011, 186, 6905–6913. [Google Scholar] [CrossRef] [Green Version]
  106. Karyampudi, L.; Lamichhane, P.; Krempski, J.; Kalli, K.R.; Behrens, M.D.; Vargas, D.M.; Hartmann, L.C.; Janco, J.M.T.; Dong, H.D.; Hedin, K.E.; et al. PD-1 Blunts the Function of Ovarian Tumor-Infiltrating Den-dritic Cells by Inactivating NF-kappa B. Cancer Res. 2016, 76, 239–250. [Google Scholar] [CrossRef] [Green Version]
  107. Zhao, Y.; Harrison, D.L.; Song, Y.; Ji, J.; Huang, J.; Hui, E. Antigen-Presenting Cell-Intrinsic PD-1 Neutralizes PD-L1 in cis to Attenuate PD-1 Signaling in T Cells. Cell Rep. 2018, 24, 379–390.e6. [Google Scholar] [CrossRef] [Green Version]
  108. Patsoukis, N.; Wang, Q.; Strauss, L.; Boussiotis, V.A. Revisiting the PD-1 pathway. Sci. Adv. 2020, 6, eabd2712. [Google Scholar] [CrossRef]
  109. Zak, K.M.; Grudnik, P.; Magiera, K.; Domling, A.; Dubin, G.; Holak, T.A. Structural Biology of the Immune Checkpoint Receptor PD-1 and Its Ligands PD-L1/PD-L2. Structure 2017, 25, 1163–1174. [Google Scholar] [CrossRef]
  110. Qin, W.T.; Hu, L.P.; Zhang, X.L.; Jiang, S.H.; Li, J.; Zhang, Z.G.; Wang, X. The Diverse Function of PD-1/PD-L Pathway Beyond Cancer. Front. Immunol. 2019, 10, 2298. [Google Scholar] [CrossRef]
  111. Pentcheva-Hoang, T.; Egen, J.G.; Wojnoonski, K.; Allison, J.P. B7-1 and B7-2 Selectively Recruit CTLA-4 and CD28 to the Immunological Synapse. Immunity 2004, 21, 401–413. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  112. Keir, M.E.; Butte, M.J.; Freeman, G.J.; Sharpe, A.H. PD-1 and Its Ligands in Tolerance and Immunity. Annu. Rev. Immunol. 2008, 26, 677–704. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  113. Karunarathne, D.S.; Horne-Debets, J.M.; Huang, J.; Faleiro, R.; Leow, C.Y.; Amante, F.; Watkins, T.S.; Miles, J.; Dwyer, P.J.; Stacey, K.; et al. Programmed Death-1 Ligand 2-Mediated Regulation of the PD-L1 to PD-1 Axis Is Essential for Establishing CD4+ T Cell Immunity. Immunity 2016, 45, 333–345. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  114. Kuipers, H.; Muskens, F.; Willart, M.; Hijdra, D.; van Assema, F.B.J.; Coyle, A.J.; Hoogsteden, H.C.; Lambrecht, B.N. Con-tribution of the PD-1 ligands/PD-1 signaling pathway to dendritic cell-mediated CD4+ T cell activation. Eur. J. Immunol. 2006, 36, 2472–2482. [Google Scholar] [CrossRef]
  115. Dulos, J.; Carven, G.J.; van Boxtel, S.J.; Evers, S.; Driessen-Engels, L.J.A.; Hobo, W.; Gorecka, M.A.; de Haan, A.F.J.; Mulders, P.; Punt, C.J.A.; et al. PD-1 Blockade Augments Th1 and Th17 and Suppresses Th2 Responses in Peripheral Blood From Patients with Prostate and Advanced Melanoma Cancer. J. Immunother. 2012, 35, 169–178. [Google Scholar] [CrossRef]
  116. Akbay, E.A.; Koyama, S.; Liu, Y.; Dries, R.; Bufe, L.E.; Silkes, M.; Alath, M.; Magee, D.M.; Jones, R.; Jinushi, M.; et al. Interleukin-17A Promotes Lung Tumor Progression through Neu-trophil Attraction to Tumor Sites and Mediating Resistance to PD-1 Blockade. J. Thorac. Oncol. 2017, 12, 1268–1279. [Google Scholar] [CrossRef] [Green Version]
  117. Liu, Y.; Cheng, Y.; Xu, Y.; Wang, Z.; Du, X.; Li, C.; Peng, J.; Gao, L.; Liang, X.; Ma, C. Increased expression of programmed cell death protein 1 on NK cells inhibits NK-cell-mediated anti-tumor function and indicates poor prognosis in digestive can-cers. Oncogene 2017, 36, 6143–6153. [Google Scholar] [CrossRef] [Green Version]
  118. Zhang, T.; Song, X.M.; Xu, L.L.; Ma, J.; Zhang, Y.J.; Gong, W.F.; Zhang, Y.L.; Zhou, X.S.; Wang, Z.B.; Wang, Y.L.; et al. The binding of an anti-PD-1 antibody to Fc gamma RI has a profound impact on its biological functions. Cancer Immunol. Immunother. 2018, 67, 1079–1090. [Google Scholar] [CrossRef] [Green Version]
  119. Tarnawski, R.; Fowler, J.; Skladowski, K.; Swierniak, A.; Suwinski, R.; Maciejewski, B.; Wygoda, A. How fast is repopula-tion of tumor cells during the treatment gap? Int. J. Radiat. Oncol. Biol. Phys. 2002, 54, 229–236. [Google Scholar] [CrossRef]
  120. Lagadec, C.; Vlashi, E.; Della Donna, L.; Dekmezian, C.; Pajonk, F. Radiation-Induced Reprogramming of Breast Cancer Cells. Stem Cells 2012, 30, 833–844. [Google Scholar] [CrossRef] [Green Version]
  121. Demicheli, R.; Retsky, M.W.; Hrushesky, W.J.M.; Baum, M.; Gukas, I.D. The effects of surgery on tumor growth: A century of investigations. Ann. Oncol. 2008, 19, 1821–1828. [Google Scholar] [CrossRef] [PubMed]
  122. Strannegård, Ö.; Thorén, F.B. Opposing effects of immunotherapy in melanoma using multisubtype interferon-alpha–can tumor immune escape after immunotherapy accelerate disease progression? OncoImmunology 2016, 5, e1091147. [Google Scholar] [CrossRef] [Green Version]
  123. Zhang, H.; Houghton, A.M. Good cops turn bad: The contribution of neutrophils to immune-checkpoint inhibitor treatment failures in cancer. Pharmacol. Ther. 2021, 217, 107662. [Google Scholar] [CrossRef] [PubMed]
  124. Sacdalan, D.B.; Lucero, J.A.; Sacdalan, D.L. Prognostic utility of baseline neutrophil-to-lymphocyte ratio in patients re-ceiving immune checkpoint inhibitors: A review and meta-analysis. Oncotargets Ther. 2018, 11, 955–965. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  125. Howard, R.; Kanetsky, P.A.; Egan, K.M. Exploring the prognostic value of the neutrophil-to-lymphocyte ratio in cancer. Sci. Rep. 2019, 9, 1–10. [Google Scholar] [CrossRef] [Green Version]
  126. Mezquita, L.; Auclin, E.; Ferrara, R.; Charrier, M.; Remon, J.; Planchard, D.; Ponce, S.; Ares, L.P.; Leroy, L.; Audigier-Valette, C.; et al. Association of the Lung Immune Prognostic Index with Immune Checkpoint Inhibitor Outcomes in Patients with Advanced Non–Small Cell Lung Cancer. JAMA Oncol. 2018, 4, 351–357. [Google Scholar] [CrossRef]
  127. Banna, G.L.; Di Quattro, R.; Malatino, L.; Fornarini, G.; Addeo, A.; Maruzzo, M.; Urzia, V.; Rundo, F.; Lipari, H.; De Giorgi, U.; et al. Neutrophil-to-lymphocyte ratio and lactate dehydrogenase as biomarkers for urothelial cancer treated with immunotherapy. Clin. Transl. Oncol. 2020, 22, 2130–2135. [Google Scholar] [CrossRef]
  128. Ferrara, R.; Russo, G.L.; Signorelli, D.; Proto, C.; Prelaj, A.; Galli, G.; de Toma, A.; Viscardi, G.; Lobefaro, R.; Brambilla, M.; et al. Circulating and tumor-associated neutrophil subtypes discriminate hyperprogressive disease (HPD) from convention-al progression (PD) upon immune checkpoint inhibitors (ICI) in advanced non-small cell lung cancer (NSCLC) patients (pts) and in vivo models. J. Clin. Oncol. 2020, 38, 9547. [Google Scholar] [CrossRef]
  129. Tesi, R. MDSC; the Most Important Cell You Have Never Heard Of. Trends Pharmacol. Sci. 2019, 40, 4–7. [Google Scholar] [CrossRef]
  130. Marvel, D.; Gabrilovich, D.I. Myeloid-derived suppressor cells in the tumor microenvironment: Expect the unexpected. J. Clin. Investig. 2015, 125, 3356–3364. [Google Scholar] [CrossRef]
  131. Groux, H.; Bigler, M.; E De Vries, J.; Roncarolo, M.G. Interleukin-10 induces a long-term antigen-specific anergic state in human CD4+ T cells. J. Exp. Med. 1996, 184, 19–29. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  132. Herbst, R.S.; Soria, J.-C.; Kowanetz, M.; Fine, G.D.; Hamid, O.; Gordon, M.S.; Sosman, J.A.; McDermott, D.F.; Powderly, J.D.; Gettinger, S.N.; et al. Predictive correlates of response to the anti-PD-L1 antibody MPDL3280A in cancer patients. Nature 2014, 515, 563–567. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  133. Hsu, J.; Hodgins, J.J.; Marathe, M.; Nicolai, C.J.; Bourgeois-Daigneault, M.C.; Trevino, T.N.; Azimi, C.S.; Scheer, A.K.; Randolph, H.E.; Thompson, T.W.; et al. Contribution of NK cells to immunotherapy mediated by PD-1/PD-L1 blockade. J. Clin. Investig. 2018, 128, 4654–4668. [Google Scholar] [CrossRef] [PubMed]
  134. Solaymani-Mohammadi, S.; Lakhdari, O.; Minev, I.; Shenouda, S.; Frey, B.; Billeskov, R.; Singer, S.; Berzofsky, J.A.; Eckmann, L.; Kagnoff, M.F. Lack of the programmed death-1 receptor renders host susceptible to enteric microbial infection through impairing the production of the mucosal natural killer cell effector molecules. J. Leukoc. Biol. 2016, 99, 475–482. [Google Scholar] [CrossRef] [Green Version]
  135. Vivier, E.; Artis, D.; Colonna, M.; Diefenbach, A.; Di Santo, J.P.; Eberl, G.; Koyasu, S.; Locksley, R.M.; McKenzie, A.N.J.; Mebius, R.E.; et al. Innate Lymphoid Cells: 10 Years On. Cell 2018, 174, 1054–1066. [Google Scholar] [CrossRef] [Green Version]
  136. Colonna, M. Innate Lymphoid Cells: Diversity, Plasticity, and Unique Functions in Immunity. Immunity 2018, 48, 1104–1117. [Google Scholar] [CrossRef] [Green Version]
  137. Castellanos, J.G.; Longman, R.S. The balance of power: Innate lymphoid cells in tissue inflammation and repair. J. Clin. Investig. 2019, 129, 2640–2650. [Google Scholar] [CrossRef] [Green Version]
  138. Chiossone, L.; Dumas, P.-Y.; Vienne, M.; Vivier, E. Natural killer cells and other innate lymphoid cells in cancer. Nat. Rev. Immunol. 2018, 18, 671–688. [Google Scholar] [CrossRef]
  139. Fung, K.Y.; Nguyen, P.M.; Putoczki, T. The expanding role of innate lymphoid cells and their T-cell counterparts in gas-trointestinal cancers. Mol. Immunol. 2019, 110, 48–56. [Google Scholar] [CrossRef]
  140. Kirchberger, S.; Royston, D.J.; Boulard, O.; Thornton, E.; Franchini, F.; Szabady, R.L.; Harrison, O.; Powrie, F. Innate lym-phoid cells sustain colon cancer through production of interleukin-22 in a mouse model. J. Exp. Med. 2013, 210, 917–931. [Google Scholar] [CrossRef]
  141. Irshad, S.; Flores-Borja, F.; Lawler, K.; Monypenny, J.; Evans, R.; Male, V.; Gordon, P.; Cheung, A.; Gazinska, P.; Noor, F.; et al. ROR gamma t+ Innate Lymphoid Cells Promote Lymph Node Metasta-sis of Breast Cancers. Cancer Res. 2017, 77, 1083–1096. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  142. Carrega, P.; Loiacono, F.; Di Carlo, E.; Scaramuccia, A.; Mora, M.; Conte, R.; Benelli, R.; Spaggiari, G.M.; Cantoni, C.; Campana, S.; et al. NCR+ILC3 concentrate in human lung cancer and associate with intratumoral lymphoid structures. Nat. Commun. 2015, 6, 8280. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  143. Xiong, D.; Wang, Y.; Singavi, A.K.; Mackinnon, A.; George, B.; You, M. Immunogenomic Landscape Contributes to Hyperprogressive Disease after Anti-PD-1 Immunotherapy for Cancer. iScience 2018, 9, 258–277. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  144. Vacca, P.; Pesce, S.; Greppi, M.; Fulcheri, E.; Munari, E.; Olive, D.; Mingari, M.C.; Moretta, A.; Moretta, L.; Marcenaro, E. PD-1 is expressed by and regulates human group 3 innate lymphoid cells in human decidua. Mucosal Immunol. 2019, 12, 624–631. [Google Scholar] [CrossRef] [PubMed]
  145. Tumino, N.; Martini, S.; Munari, E.; Scordamaglia, F.; Besi, F.; Mariotti, F.R.; Bogina, G.; Mingari, M.C.; Vacca, P.; Moretta, L. Presence of innate lymphoid cells in pleural effusions of primary and metastatic tumors: Functional analysis and expression of PD-1 receptor. Int. J. Cancer 2019, 145, 1660–1668. [Google Scholar] [CrossRef]
  146. Hepworth, M.R.; Fung, T.C.; Masur, S.H.; Kelsen, J.R.; McConnell, F.M.; Dubrot, J.; Withers, D.R.; Hugues, S.; Farrar, M.A.; Reith, W.; et al. Group 3 innate lymphoid cells mediate intes-tinal selection of commensal bacteria-specific CD4(+) T cells. Science 2015, 348, 1031–1035. [Google Scholar] [CrossRef] [Green Version]
Figure 1. Zappasodi et al. illustrated the function of 4PD1hi cells (PD-1+CD4+Foxp3 T cells) and observed that these cells accumulate within the tumor as a function of the tumor burden. Reprinted with permission from Ref. [61]. 2018, Elsevier.
Figure 1. Zappasodi et al. illustrated the function of 4PD1hi cells (PD-1+CD4+Foxp3 T cells) and observed that these cells accumulate within the tumor as a function of the tumor burden. Reprinted with permission from Ref. [61]. 2018, Elsevier.
Cells 11 01758 g001
Figure 2. Hypothesized mechanism through which macrophages and ICIs are involved in HPD development. Reprinted with permission from Ref. [10]. 2019, American Association for Cancer Research.
Figure 2. Hypothesized mechanism through which macrophages and ICIs are involved in HPD development. Reprinted with permission from Ref. [10]. 2019, American Association for Cancer Research.
Cells 11 01758 g002
Figure 3. (A) Anti-PD-L1 antibodies (IgG1) and anti-PD-1 antibodies (IgG4) have different molecular mechanisms exerted through binding with activating or inhibitory FcRs. (B) FcyRIIB-enhanced clustering of PD-1 on macrophages results in polarization via enhanced signaling based on the immunoreceptor tyrosine-based inhibitory motif (ITIM) domain. Reprinted with permission from Ref. [100]. 2019, American Association for Cancer Research.
Figure 3. (A) Anti-PD-L1 antibodies (IgG1) and anti-PD-1 antibodies (IgG4) have different molecular mechanisms exerted through binding with activating or inhibitory FcRs. (B) FcyRIIB-enhanced clustering of PD-1 on macrophages results in polarization via enhanced signaling based on the immunoreceptor tyrosine-based inhibitory motif (ITIM) domain. Reprinted with permission from Ref. [100]. 2019, American Association for Cancer Research.
Cells 11 01758 g003
Table 1. Representative case series studies of HPD in multiple cancer types.
Table 1. Representative case series studies of HPD in multiple cancer types.
ReferenceYearCountryCancer TypesIncidence, n (%)Clinical Relevance
Champiat et al. [6]2017FrancePan-cancer12/131 (9%)HPD was associated with a higher age (>65 years old) and a worse OS, but not associated with higher tumor burden or any specific tumor type.
Saâda-Bouzid et al. [7] 2017FranceR/M HNSCC10/34 (29%)HPD significantly correlated with a regional recurrence, but not with local or distant recurrence. HPD was associated with a shorter PFS, but not with OS.
Ferrara et al. [8]2018FranceNSCLC56/406 (13.8%)HPD was associated with high metastatic burden and poor prognosis.
Kanazu et al. [9]2018Japan NSCLC 5/87 (5.7%)HPD was thought to be associated with poor quality of life and survival.
Lo Russo et al. [10]2019ItalyNSCLC39/152 (25.7%)Pretreatment tissue samples from all patients with HPD showed tumor infiltration by M2-like CD163+CD33+PD-L1+ clustered epithelioid macrophages.
Kim et al. [11]2019South KoreaR/M NSCLC55/263 (20.9%)A lower frequency of effector/memory subsets (CCR7CD45RA T cells among the total CD8+ T cells) and a higher frequency of severely exhausted populations (TIGIT+ T cells among PD-1+CD8+ T cells) in peripheral blood were associated with HPD and inferior survival rate.
Aoki et al. [12]2019JapanAGC19/100 (19.0%)HPD was observed more frequently after nivolumab compared with irinotecan, which was associated with a poor prognosis after nivolumab but not so clearly after irinotecan.
Kanjanapan et al. [13]2019CanadaPan-cancer12/182 (6.6%)HPD was not associated with CSAEs, age, tumor type, or the type of immunotherapy but was more common in females.
Sasaki et al. [14]2019JapanAGC13/62 (21.0%)Elevations in ANC and CRP levels upon nivolumab treatment might indicate HPD.
Lu et al. [15] 2019ChinaMetastatic GTC5/56 (8.9%)Baseline serum levels of MCP-1, LIF, and CD-152 were associated with HPD.
Matos et al. [16]2020SpainPan-cancer29/270 (10.7%)The HPD progressor group had a significantly lower OS when compared with the non-HPD progressor group.
Castello et al. [17]2020ItalyNSCLC14/46 (30.4%)HPD status was associated with tumor burden. The derived neutrophil-to-lymphocyte ratio and platelet count were significantly associated with HPD status.
Hwang et al. [18]2020South KoreaRCC/UC13/203 (6.4%)HPD developed predominantly in patients with UC, and the incidence of HPD in patients with RCC was negligible. UC and creatinine above 1.2 mg/dL were independent predictive factors for HPD. A 30% increase in lymphocyte number following PD-1/PD-L1 inhibitor treatment was a negative predictor of HPD.
Vaidya et al. [19]2020USAAdvanced NSCLC19/109 (17.4%)Image-based radiomics markers extracted from baseline CTs might help identify patients at risk of HPD.
Zhang et al. [20]2020ChinaAdvanced HCC10/69 (14.5%)Haemoglobin level, portal vein tumour thrombus, and Child-Pugh score were significantly associated with HPD. Patients with HPD had a significantly shorter OS than that of the patients with non-HPD.
Kim et al. [21]2021South KoreaAdvanced HCC24/189 (12.7%)Patients with HPD had worse PFS and OS compared to patients with progressive disease without HPD. An elevated neutrophil-to-lymphocyte ratio (>4.125) was associated with HPD and an inferior survival rate.
Chen et al. [22]2021ChinaPan-cancer38/377 (10.1%)Patients with HPD had lower OS than those without HPD. KRAS status was significantly associated with HPD in patients with colorectal cancer. The rapid increase of characteristic tumor markers within 1 month was associated with the occurrence of HPD.
Xiao et al. [23]2021ChinaPLC13/129 (10.1%)The PFS of HPD patients was significantly worse than that of non-HPD patients. Compared with the non-HPD patients, lung metastasis, and lymph node metastasis were independent risk factors of HPD.
Miyama et al. [24]2021JapanUC6/23 (26.1%)Squamous differentiation may be a novel biomarker for predicting HPD in patients with UC who receive pembrolizumab.
Maesaka et al. [25]2022JapanUnresectable HCC9/88 (10.2%)Patients with HPD had larger and more intrahepatic tumors, higher levels of α-fetoprotein and lactate dehydrogenase, and higher NLR at baseline than patients without HPD. NLR of ≥3 at baseline was identified as the only independent factor associated with HPD in multivariate analysis.
R/M, recurrent, and/or metastatic; HNSCC, head, and neck squamous cell carcinoma; NSCLC, non-small cell lung cancer; AGC, advanced gastric cancer; GTC, gastrointestinal tract cancer; RCC, renal cell carcinoma; UC, urothelial carcinoma; HCC. hepatocellular carcinoma; PLC, primary liver cancer; OS, overall survival; PFS, progression-free survival; CSAEs, clinically significant adverse events; ANC, absolute neutrophil count; CRP, C-reactive protein; MCP-1, monocyte chemoattractant protein 1; LIF, leukemia inhibitory factor; CD-152, cluster of differentiation 152; NLR, neutrophil-to-lymphocyte ratio.
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Share and Cite

MDPI and ACS Style

Wei, Z.; Zhang, Y. Immune Cells in Hyperprogressive Disease under Immune Checkpoint-Based Immunotherapy. Cells 2022, 11, 1758. https://doi.org/10.3390/cells11111758

AMA Style

Wei Z, Zhang Y. Immune Cells in Hyperprogressive Disease under Immune Checkpoint-Based Immunotherapy. Cells. 2022; 11(11):1758. https://doi.org/10.3390/cells11111758

Chicago/Turabian Style

Wei, Zhanqi, and Yuewei Zhang. 2022. "Immune Cells in Hyperprogressive Disease under Immune Checkpoint-Based Immunotherapy" Cells 11, no. 11: 1758. https://doi.org/10.3390/cells11111758

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop