Next Article in Journal
Mitochondrial Role in Oncogenesis and Potential Chemotherapeutic Strategy of Mitochondrial Infusion in Breast Cancer
Next Article in Special Issue
The Interplay of Type 1, Type 2, and Type 3 Lymphocytes and Cytokines in Atopic Dermatitis
Previous Article in Journal
Microglia and Alzheimer’s Disease
Previous Article in Special Issue
Editorial: Special Issue, “Molecular Advances in Skin Diseases”
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Changes in the Immune Cell Repertoire for the Treatment of Malignant Melanoma

Department of Dermatology, Shinshu University School of Medicine, Matsumoto 390-8621, Japan
*
Author to whom correspondence should be addressed.
Int. J. Mol. Sci. 2022, 23(21), 12991; https://doi.org/10.3390/ijms232112991
Submission received: 14 October 2022 / Accepted: 26 October 2022 / Published: 27 October 2022
(This article belongs to the Special Issue Molecular Advances in Skin Diseases 2.0)

Abstract

:
Immune checkpoint inhibitors (ICIs) have been used for the treatment of various types of cancers, including malignant melanoma. Mechanistic exploration of tumor immune responses is essential to improve the therapeutic efficacy of ICIs. Since tumor immune responses are based on antigen-specific immune responses, investigators have focused on T cell receptors (TCRs) and have analyzed changes in the TCR repertoire. The proliferation of T cell clones against tumor antigens is detected in patients who respond to treatment with ICIs. The proliferation of these T cell clones is observed within tumors as well as in the peripheral blood. Clonal proliferation has been detected not only in CD8-positive T cells but also in CD4-positive T cells, resident memory T cells, and B cells. Moreover, changes in the repertoire at an early stage of treatment seem to be useful for predicting the therapeutic efficacy of ICIs. Further analyses of the repertoire of immune cells are desirable to improve and predict the therapeutic efficacy of ICIs.

1. Introduction

Immune checkpoint inhibitors (ICIs) are currently used for the treatment of various types of cancers, including malignant melanoma [1]. However, only approximately 30–50% of melanoma patients respond to treatment with ICIs; thus, their therapeutic efficacy needs to be improved [2,3]. The analysis of “tumor antigen-specific immune responses”, which represent an important part of immune responses to tumors, can aid in improving the therapeutic efficacy of ICIs [4,5]. Therefore, changes in the repertoires of immune cells that recognize tumor antigens have been analyzed [6]. Multiple studies have reported that the ICI-induced restoration of exhausted antitumor immune responses allows for the clonal proliferation of immune cells and produces therapeutic benefits [7,8,9]. In this article, we review recent progression on the repertoires of various immune cells and their associations with treatment and symbiotic bacteria in malignant melanoma.

2. T Cells and Their Repertoires

2.1. T Cell Receptor (TCR)

T cells are activated by the recognition of corresponding antigens via TCRs. Most T cells express αβTCR, a heterodimer of TCRα and TCRβ chains, whereas some T cells express γδTCR, a heterodimer of TCRγ and TCRδ chains. T cells expressing αβTCR are referred to as αβT cells, whereas T cells expressing γδTCR are referred to as γδT cells.
There are between 2 × 109 and 6 × 109 unique TCRs in the circulating T cell population of a healthy individual [10]. Such a large number of unique clonotypes are stochastically generated via V(D)J recombination in thymic T cell progenitors, followed by positive and negative selection, which select T cells that bind to major histocompatibility complex (MHC) but not to self-peptides. TCRs are responsible for the sensitivity and specificity of T cells to peptides presented by MHC [11]. Once the TCR binds to a peptide, signal transduction occurs, which promotes activation, proliferation, differentiation, cytokine production, and an increase in cytolytic activity [12]. Through unique TCRs on individual T cells, T cells can scan antigens presented by MHC molecules on the surface of tumor cells [13]. The recognition of an antigen by a TCR results in the activation and proliferation of antigen-reactive T cells, and a process of clonal proliferation is triggered [14].
Tumor-specific T cells develop as a response to tumor antigens, including individual “neoantigens” derived from mutant proteins of cancer cells [15]. The large variety of TCRs required for effective immune function is produced via error-prone recombination of the TCR loci [16,17]. In particular, complementarity determining region 3 (CDR3) of the TCR is highly diversified and has sequences that are unique to individual T cell clones, and T cell responses can be traced by sequencing the CDR3 regions of TCRs. The sequencing efforts have been focused particularly on CD8-positive T cells, which play a central role in immune responses to tumors. The therapeutic efficacy of ICIs is correlated with the proliferation of T cell clones possessing TCRs that recognize tumor antigens (decrease in the T cell repertoire size) [7]. The therapeutic efficacy of ICIs and changes in the repertoire are described in detail later.

2.2. CD8-Positive T Cells

CD8-positive T cells recognize tumor antigens via MHC class I and play a major role in tumor immunity. Therefore, the tumor-infiltrating CD8-positive T cell count is a prognostic factor in patients with cancer [18].
Tumor-infiltrating lymphocytes (TILs) form a polyclonal population in which T cells targeting known and unknown tumor-specific antigens, such as tumor-associated antigens and neoantigens, are enriched [18]. A recent study showed a relatively large overlap between the TCR repertoire of TILs and that of CD8-positive PD-1 (programmed cell death 1)-positive T cells in the peripheral blood [19]. Furthermore, the TCR repertoire of peripheral blood PD-1-positive lymphocytes proliferated in vitro is highly similar to that of TILs [20]. Treatment with these proliferated lymphocytes administered concomitantly with an anti-PD-1 antibody led to successful outcomes in melanoma patients [7].
Furthermore, the TCR repertoire in melanoma patients before starting immunotherapy was compared with that in healthy individuals [21]. The TCR repertoire of CD8-positive T cells in the blood of melanoma patients was significantly more restricted than that in healthy individuals. In melanoma patients, the repertoire of CD8-positive T cells in the peripheral blood was more restricted than that of CD4-positive T cells. In addition, the same CD8-positive T cell clones were enriched in the peripheral blood and tumors after treatment with an anti-CTLA-4 (cytotoxic T-lymphocyte-associated protein 4) antibody. In another study, T cell clones present in both tumors and peripheral blood were investigated to analyze their ongoing immune responses [22].
A single-cell level analysis of transcriptome and TCRαβ repertoire pairs was conducted with peripheral blood T cells and tumor-infiltrating T cells from matched patients with metastatic melanoma. The effector function gene signatures in clonally proliferated TILs were similar to those in T cells in matched peripheral blood. In contrast, exhaustion factors (e.g., PD-1) were expressed mainly on tumor-infiltrating T cells. Moreover, significantly more peripheral blood T cells possessing a tumor antigen-recognizing repertoire were found in tumor tissues after immunotherapy. These data suggest that peripheral blood T cells are useful for assessing T cell function in cancer immunotherapy.
Interestingly, clinical responses to immunotherapy are associated with an influx of novel T cell clones from the peripheral blood into the tumor microenvironment [8]. In other words, peripheral blood T cells are supplied as TILs into the tumor and are involved in tumor rejection. T cells primed locally in tumors are more likely to express exhaustion factors (e.g., PD-1) than peripheral blood T cells. In fact, some antigen-presenting cells (APCs) are unsuitable for priming T cell responses in tumors as a result of interactions with regulatory (Treg) T cells [23].

2.3. CD4-Positive T Cells

CD4-positive T cells recognize tumor antigens via MHC class II and assist CD8-positive T cells in antitumor immunity [24]. Maintenance of CD4-positive T cells depends on the presence of APCs, which include B cells, macrophages, and dendritic cells (DCs). CD4-positive T cells typically do not have a cytotoxic function; however, their role in cancer has been revisited recently owing to the fact they are required for proper activation of immune responses and maintenance of immunological memory [25]. The pretreatment TCR repertoire of CD4-positive T cells in melanoma patients is significantly more restricted than that in healthy individuals [22]. Moreover, CD4-positive T cell clones in peripheral blood proliferate in melanoma patients after treatment with an anti-CTLA-4 antibody, and these CD4-positive T cell clones are enriched in corresponding tumors. In addition, CD4-positive T cell clones are less proliferative than CD8-positive T cell clones [26]. Investigation of tertiary lymphoid structures in human cancer may be a feasible approach to analyze the immune responses of CD4-positive T cells.

2.4. Regulatory T Cells

CD4-positive Treg cells play important roles in the maintenance of self-tolerance [27]. However, they play a negative role in antitumor immune responses [28]. In contrast to Treg cells circulating in the blood, Treg cells present in tumors have a profile of activated cells [29]. Increases in the number of Treg cells in tumors are correlated with disease progression and decreased survival in patients with cancer. However, the antigen specificity of Treg cells that undergo proliferation in the tumor microenvironment has not been fully understood. Thus, the TCR profiles of intratumoral Treg cells were analyzed to elucidate their antigen specificity [30]. The TCR repertoire of Treg cells in tumors varied but had significant overlap with that of Treg cells in peripheral blood. In contrast, the TCR repertoire of Treg cells had no overlap with that of conventional T cells in tumors or peripheral blood. The TCRs of Treg cells seem to have specific reactivity to tumor antigens and neoantigens. These findings suggest that intratumoral Treg cells affect the activation and clonal proliferation of T cells in a tumor antigen-selective manner. In addition, tumor antigen-specific Treg-derived TCRs have been detected in tumors and peripheral blood, suggesting that Treg cells in peripheral blood serve as a source of tumor-specific TCRs.

2.5. Effector Memory T (TEM) Cells, Central Memory T (TcM) Cells, and Resident Memory T (TRM) Cells

While recent research has focused on T cell responses to cancer immunotherapy, long-term immunological memory has not been analyzed fully. In general, some effector T cells differentiate into central memory T (TCM) cells and effector memory T (TEM) cells. TCM cells express CCR7 and CD62L and are found in secondary lymphoid tissues. TEM cells circulate in secondary lymphoid and peripheral tissues. TEM and TCM cells are chiefly found among CD8-positive memory T cells in the tumor microenvironment [31].
PD-1, an immune checkpoint protein, is expressed in effector, effector memory, and central memory CD8-positive T cells in addition to exhausted T cells [32]. Among TILs, the presence of TCM cells serves as a positive predictor of response to immunotherapy [33]. This may be because TCM cells are capable of expanding during ICI therapy.
TCM cells are characterized by the transcription factors TCF7 and T-bet. These are essential for central memory and maintenance of more differentiated T cells, respectively. An analysis conducted in melanoma patients showed that T-bet and TCF7 are upregulated by BRAF/MEK inhibition [34]. The repertoire of T cells found in tumors before treatment is expanded after the administration of BRAF/MEK inhibitors. TCF7 is involved in the self-renewal and persistence of CD8-positive memory T cells [35]. T-bet is important to maintain the balance between T cell memory and effector differentiation pathways and to trigger Th1 responses characterized by the interferon gamma (IFN-γ) signature [36]. These findings suggest that TCF7 and T-bet induced during BRAF/MEK inhibition promote the repertoire expansion of intratumoral T cells [34].
Recently, resident memory T (TRM) cells have been discovered as a subset of long-lived memory T cells. TRM cells express surface markers such as CD103 and reside in peripheral nonlymphoid tissues [37,38]. They play a role in immune responses to microbial infection in barrier tissues, such as the skin. It is likely that the presence of TRM cells is associated with improved long-term prognosis in various types of cancers, including melanoma [39,40,41]. TRM cells have been detected in tumors before treatment [42].
In addition, memory CD8-positive T cell responses were investigated in melanoma patients who showed a long-term response (for at least 1 year) to immunotherapy [43]. Single-cell RNA sequencing revealed TRM cells that were shared between tumors and the vitiliginous skin. TCR sequence analysis identified clonotypes that coexisted as TRM cells in the skin and as TEM cells in the blood, and clonotypes found in tumors, skin, and peripheral blood expressed high levels of IFNγ and TNFα. Higher levels of these cells were strong predictors of favorable prognosis in melanoma patients. Intratumoral clonotypes were found in the skin and blood of patients for up to 9 years, and the tumor-related clone repertoire was abundantly maintained in the skin. In the future, analyses of the effects of TRM and TEM cells on the clonal proliferation of T cells by immunotherapy are anticipated.

3. B Cells and Their Repertoires

3.1. B Cell Antigen Receptors (BCRs)

BCRs are expressed on the surface of B cells and bind directly to antigens. When two or more BCRs recognize a multivalent antigen, the BCRs becomes cross-linked and signal transduction ensues [44]. This results in the proliferation, differentiation, and activation of B cells. Antigens bound to the BCRs are taken up by the cells and degraded into peptides, which are then presented by MHC class II. The antigen-presenting B cells interact with T cells that are reactive to the same antigens, inducing the formation of a germinal center and differentiation of plasma cells. In addition, antigen-recognizing B cells do not require the help of T cells for proliferation and differentiation into plasma cells.

3.2. B Cells

In melanoma, tumor-associated B cells account for up to 33% of immune cells [45]. Multiple studies on the role of tumor-associated B cells have been reported recently. The CD27-positive, CD38-positive, and PAX5-negative tumor-induced plasmablast-like-enriched B cell (TIPB) population regulates T cell-recruiting chemokines (CCL3, CCL4, and CCL5) and increases the number of tumor-infiltrating T cells. ICIs seem to be more effective in cases with a larger TIPB population [46], and tumor-associated B cells are related with improved survival independent of other clinical variables. This result may be explained by the induction of tertiary lymphoid structures [47,48]. Immunofluorescence staining of CD20, CXCR5, and CXCL13 revealed the formation of tertiary lymphoid structures in tumors. Interestingly, naïve B cells and plasma cells were more abundant in responders at baseline, and a higher number of intratumoral memory B cells was associated with a more diverse TCR repertoire. Furthermore, a higher number of plasma cells was associated with a higher number of infiltrating T cells. Among melanoma patients treated with ipilimumab and nivolumab, those with a larger number of immunoglobulin gene rearrangements in the tumors exhibited significantly longer progression free survival. Therefore, the intratumoral B cell repertoire should be evaluated to understand B cell immunity. The CDR3 sequences of B cell immunoglobulin heavy chain (IgH) and light chain (IgL) were assembled from bulk RNA-sequence data to analyze the function of intratumoral B cells in antitumor immune responses [49]. Clones of both IgH and IgL chains were significantly increased in number in tumors of patients who responded to treatment with ICIs. It is likely that the abundance of BCR clonotypes in tumors at baseline is positively correlated with the therapeutic efficacy of ICIs. The tumors of patients who responded to the treatment tend to have more clonal BCR repertoire.

4. Treatment and Repertoires

4.1. Changes in the Repertoire with ICIs

PD-1 blockade restores T cell functions primarily at the effector phase. Therefore, PD-1 blockade rescues exhausted CD8-positive T cells and restores their cytotoxic capacity, thereby facilitating the destruction of tumor cells [50]. Anti-PD-1 antibodies prevent PD-1 from binding to PD-L1 on APCs and interfering with the differentiation and functioning of Treg cells because PD-1 plays a role in the development of Treg cells [51]. However, the PD-1 expression pattern encompasses a broad range of immune cells, including T cells, B cells, natural killer cells, dendritic cells, and bone marrow cells. Its ligands, PD-L1 (programmed death-ligand 1) and PD-L2, are also expressed in various hematopoietic and nonhematopoietic cells as well as cancer cells [52,53]. Therefore, anti-PD-1 therapy can affect various types of cells and pathways.
Recent analyses have mainly focused on CD8-positive T cells, which play a major role in tumor immune responses. After anti-PD-1 therapy, most of the abundant TIL clones were found among T cell clones in peripheral blood, regardless of the clinical response [9]. T cell clones expressing TCRs that recognize tumor antigens proliferate in anti-PD1 antibody responders [7,54]. Moreover, an analysis in patients who underwent neoadjuvant anti-PD-1 antibody therapy revealed a post-treatment increase in the number of CD8-positive T cells reactive with gp100, a melanoma-specific antigen [55]. These findings suggest that the ICI-induced restoration of exhausted antitumor immune responses is followed by the clonal proliferation of immune cells, yielding therapeutic benefits.
Meanwhile, blockade of CTLA-4, another immune checkpoint protein, decreases the T cell priming threshold and allows for the proliferation of more effector T cells [56]. Anti-CTLA-4 antibodies also allow for the proliferation of memory T cell clones [57]. Moreover, Treg cells in the tumor microenvironment are depleted as they express CTLA-4 [58]. Depletion of Treg cells improves intratumoral IL-2 levels, facilitates survival of CD8-positive T cells, and expands the TCR repertoire [59]. In other words, anti-CTLA-4 antibodies inhibit Treg cells, reverse inhibit CD8-positive T cells, and expand the TCR repertoire in a nonspecific manner [60]. In addition, anti-CTLA-4 antibodies accelerate the turnover of T cell repertoire and increase TCR diversity [61,62,63]. Anti-CTLA-4 antibodies broaden the T cell repertoire, whereas anti-PD1 antibodies promote the proliferation of a limited number of clones, skewing the T cell repertoire (Figure 1).
A previous study showed tumor antigenic changes after PD-1 blockade [64]. Thus, a dynamic clonal change in the TIL could be induced by antigenic alteration of tumor cells during treatment [64], whereas the clone of TIL expanded after PD-1 blockade responded to tumor cell line before PD-1 blockade [65]. Therefore, the authors reported that dynamic tumor-specific clonal changes after PD-1 blockade are caused by PD-1 blockade but not antigenic alteration of tumor cells [65].
T cells use TCRs to recognize antigen peptides presented by MHC on antigen-presenting cells. CD28 receives stimulus from CD80/86, which leads to T cell activation. CTLA-4 competes with CD28 and inhibits T cell activation. Therefore, inhibition of CTLA-4 results in nonspecific proliferation of T cell clones and expansion of the TCR repertoire. On the other hand, T cells use TCRs to recognize antigen peptides on tumor cells for eliciting antitumor immune responses, and tumor PD-L1 binding to PD-1 on T cells inhibits the antitumor immune responses. PD-1 inhibition promotes the proliferation of specific T cell clones, leading to alteration of the TCR repertoire.

4.2. Changes in the Repertoire with BRAF/MEK Inhibition

MEK inhibitors can impair T cell activation, because T cell activation mediated by TCRs and their co-stimulatory molecules is dependent on mitogen-activated protein kinase (MAPK) and the PI3K-AKT signaling cascade [66]. In fact, pharmacological in vitro inhibition of MEK had adverse effects on T cell activation [67].
In contrast, the results of an in vivo analysis were twofold: MEK inhibition had no adverse effects on T cell effector function and showed favorable outcomes in combination with immune checkpoint inhibition [68]. MEK inhibition was associated with increased tumor-infiltrating CD8-positive T cells, increased IFN-γ gene expression signatures, and decreased abundance of tumor-associated macrophages and Treg cells. Furthermore, MEK inhibition protected effector T cells from activation-induced cell death due to chronic TCR stimulation [69]. In addition, an analysis in melanoma patients showed that BRAF/MEK inhibition upregulated the levels of T-bet and TCF7 and expanded T cell repertoire in tumors [34]. Another study showed that higher intratumoral T cell clonality was associated with better responses to BRAF inhibitor treatment for melanoma [70]. Based on these results, efficacy analyses of ICIs combined with BRAF/MEK inhibitors are desired in clinical trials.

4.3. Changes in the Repertoire with Adoptive Cell Transfer of TIL Therapy

Adoptive cell transfer (ACT) of TIL is used for the treatment of advanced melanoma. ACT of TIL has shown significant clinical benefit [71]. However, ACT of TIL could not work enough in patients previously treated with PD-1 or MAPK inhibition [68]. Response to ACT correlates with the recognition of tumor neo-antigens [72,73]. Anti-PD-1 naïve patients were received TIL reactive with more neo-antigens compared with anti-PD-1 experienced patients [74]. Treatment products administered to anti-PD-1 naïve patients were more likely to contain T cells reactive against neoantigens than treatment products for anti-PD-1 experienced patients [74].

4.4. Changes in the Repertoire with IL-12 Therapy

IL-12, an inflammatory cytokine, induces the proliferation and activation of natural killer (NK) cells and cytotoxic T cells and enhances effector functions [75,76]. Additionally, it is an important link between innate and acquired immune systems, because APC-producing IL-12 stimulates the release of IFN-γ from T cells and NK cells [77]. It is also involved in Th1 induction and promotes IFN-γ production [78]. Thus, IL-12 plays a role in antitumor immunity, and T cells are important for IL-12-mediated tumor suppression [79]. Intratumoral plasmid IL-12 electroporation therapy was tested in a phase II trial in melanoma patients [80]. Following the treatment, intratumoral T cells proliferated clonally, which led to a skewed TCR repertoire.

4.5. Treatment Correlations and Repertoires

CTLA-4 is expressed mainly on CD4-positive T cells after TCR-mediated activation and interferes with CD28 co-stimulatory signaling induced by APCs to inhibit TCR-induced activation and proliferation [81]. Therefore, CTLA-4 inhibition induces the activation and proliferation of antitumor T cells via increased CD28 signaling.
PD-1 inhibits the effector function of antigen-specific T cells upon binding to ligands [82]. PD-1 inhibitors directly regulate the functions of various types of PD-1-expressing immune cells [83]. Immunotherapy with anti-PD-1 antibodies is widely used for the treatment of metastatic solid tumors, with a response rate of 20–55% [3]. Biomarkers to predict which patients are likely to respond to anti-PD-1 antibody therapy are needed.
Because highly variable CDR3 of the TCR chain is unique to individual T cell clones, CDR3 can be used to monitor the dynamics of T cell repertoire responses to ICIs [84]. A recent study showed that the TCR clonality or diversity of T cells in the blood increases 3 weeks after the initiation of treatment with anti-PD-1 antibodies [7]. Clonal proliferation of TCRs in the blood also occurred only in responders 3 weeks after the initiation of combination treatment with anti-PD-1 and anti-CTLA-4 antibodies. Thus, with this approach, minimally invasive liquid biopsies may be used in the early stages of treatment to predict patients’ treatment response.
The anti-PD-1 antibody treatment seems to be more effective in melanoma patients when the pretreatment TCR repertoire of TILs is larger in metastatic tumors [85]. An analysis of melanoma in The Cancer Genome Atlas reveals that a larger TCR repertoire of TILs is associated with longer overall survival even without anti-PD-1 antibody treatment. Furthermore, TCR repertoires in the peripheral blood of melanoma patients were examined to determine whether the TCR diversity predicted the clinical prognosis of ICI treatment [20]. Higher TCR repertoire diversity in the blood was associated with longer progression free survival, and low repertoire diversity was associated with poor prognosis. The diversity in patients who experienced late recurrence and long-term survival was significantly higher than that in rapid progressors. The TCR repertoire diversity in tumors may have a potential prognostic value.
Liquid biopsies were performed before treatment to predict responses to ICIs [86]. In the PCR analysis of pretreatment peripheral blood mononuclear cells (PBMCs), the diversity evenness of the TCRs repertoire score was correlated with the therapeutic efficacy of ICIs. Furthermore, the pretreatment level of TCR repertoire restrictions in CD4-positive T cells in peripheral blood had a potential prognostic value for clinical response to CTLA4 inhibition [20]. In addition, T cells release their DNA into the blood when cell death occurs. The released cell-free DNA in the blood was sequenced for analyzing CDR3 of T cells [63], which suggested that the clonal proliferation of T cell repertoire in the blood within 3 weeks of starting ICI treatment predicts the therapeutic efficacy.

4.6. Immune-Related Adverse Events and Repertoires

Anti-CTLA-4 and anti-PD-1 therapies can prolong survival in melanoma patients; however, these therapies can also induce organ-specific toxicities, called immune-related adverse events (IrAEs), which make it impossible to continue ICIs in a considerable number of patients [7,87]. It is likely that T cell clonal analysis is useful for the early diagnosis of IrAEs. The number of T cell clones that newly underwent proliferation was higher in patients with severe IrAEs after CTLA-4 blockade [88]. Furthermore, newly expanded clones were found among CD8-positive T cells, but not CD4-positive T cells, in patients with IrAEs [89]. In addition, severe repertoire restrictions were found in CD4-positive T cells in a study using samples of severe colitis [22]. CD4-positive and CD8-positive T cells may be diversely involved in various IrAEs; further studies are necessary for clarifying this point.

4.7. Repertoire Analysis and Vaccines

Peptides recognized by TCRs are used as vaccines to enhance antitumor immune responses [90]. Specifically, tumor biopsy specimens and nonmalignant tissue samples (usually PBMCs) are collected from patients and subjected to whole exome sequencing for comparison between tumor DNA and germline DNA to identify tumor-specific somatic mutations. A computational approach is used to predict MHC class I-binding epitopes, and peptides predicted to have moderate-to-high MHC-binding affinities are likely to induce CD8-positive T cell responses [91].
MHC class II-binding peptides are often more difficult to predict. The peptide-binding groove of MHC class I has closed ends to define the arrangement of a peptide epitope composed of 8–11 amino acid residues for presentation to CD8-positive T cells. In contrast, the peptide-binding groove of MHC class II has open ends and can bind to a peptide that is longer and more variable in length. Recently, new methods have been developed for the prediction of MHC class II-binding peptides [92,93,94], and they are expected to promote the development of MHC class II-binding peptides to elicit tumor-specific responses of CD4-positive T cells.

5. Repertoires and Tumor Heterogeneity

Relationships between TILs and melanoma during ICI therapy have been analyzed [95]. In responders to ICIs, the proportion of neoantigen-expressing tumors was reduced after the treatment, and tumor-infiltrating T cell clones expressing TCRs that recognize neoantigens were increased inversely. This result suggests that the proliferated neoantigen-recognizing T cell clones remove neoantigen-expressing tumor cells in responders to ICI therapy. Conversely, in nonresponders, clones with new mutations emerge during the treatment.
Repertoire analysis of CD8-positive T cells from tumors at different sites has also been performed [96]. Common CD8-positive T cell clones were found in tumors at different sites; however, the number of CD8-positive T cells for those clones varied widely depending on the tumor site. Because tumors at different sites express various tumor antigens in different ratios, CD8-positive T cell counts for clones should differ depending on the tumor site.

6. Symbiotic Microorganisms and Repertoires

6.1. Intestinal Bacteria

The compositions of intestinal bacteria in responders to ICIs are different from those in nonresponders [97,98]. Intestinal bacterial flora affects the innate and adaptive immune systems, thereby regulating the effectiveness of various antitumor treatments [99]. A recent study revealed how antigen mimicry between symbiotic bacteria and tumor antigens induces beneficial antitumor responses [100]. In a mouse model, Bifidobacterium spp., whose components have antigenic homologies to neoantigens of mouse melanoma, stimulate tumor-specific T cells in the form of cross-reactivity. Bifidobacterium spp. have also been shown to induce clonal proliferation of T cell repertoires that recognize neoantigens. In fact, fecal microbiota transplantation increased the tumor immunity [101]. Collectively, homologies between tumor neoantigens and pathogens or symbiotic bacteria may provide an effective method for isolating a unique T cell subpopulation with a potent antitumor function.

6.2. Intracellular Bacteria

Various bacteria are known to colonize human tumors [102]. Tumor-associated bacteria may affect immune function, treatment responses, and patient survival [103]. However, it remains to be fully elucidated whether antigens derived from intracellular bacteria are presented by MHC class I and II of tumor cells or whether such antigens induce TILs.
Recently, 16S rRNA gene sequencing was used to analyze intratumoral bacteria in melanoma [104], which found 41 bacterial species in 17 metastatic melanomas. High similarities in the composition of the identified bacteria were shown among samples from different patients, which suggested the presence of common bacterial species in melanoma cells. MHC peptidomics analyses have revealed 248 and 35 unique class I and class II peptides derived from 41 bacterial species. In addition, peptides derived from intracellular bacteria may stimulate CD8-positive T cells via MHC class I on tumor cells. Extracellular bacterial proteins taken up by tumors may stimulate CD4-positive T cells via MHC class II on tumor cells [105]. Bacterial antigens will be useful targets for immunotherapy as nonself antigens. In fact, TILs involved in the identification of bacteria-derived antigens were shown to secrete IFNγ and express CD69.

6.3. Viruses

CD8-positive T cells play a role in the detection and removal of cells presenting abnormal peptides on the surface as a result of viral infection or malignant transformation. Interestingly, antiviral T cells have been detected in the tumor microenvironment [106]. Tumor-specific peptides are occasionally similar to virus-derived peptides [107]. PBMCs were collected from melanoma patients before and after anti-PD-1 antibody treatment, and the cross-reactivity of PBMCs to tumors and cytomegalovirus (CMV) peptides was assessed. PBMCs were stimulated by both tumor peptides and CMV homologs, which suggests that CMV peptides expand virus-specific T cell clones capable of attacking and killing melanoma cells.
Meanwhile, CD8-positive memory T cells that do not recognize tumor antigens; however, these cells can recognize EB virus antigens, have been reported to account for 22% of TILs [108]. TILs recognizing tumor antigens can be distinguished from such non-tumor-reactive TILs based on higher levels of TCF7 transcription, PD-1 expression, and CD39 expression.

7. Summary

Analysis of immune cell repertoires has led to the elucidation of antitumor immune responses to melanoma. In particular, repertoire changes are important to predict the therapeutic efficacy of ICIs. The knowledge acquired through repertoire analysis of immune cells can be applied for enhancing tumor immune responses to improve the prognosis of melanoma. Currently, methods to enhance the therapeutic efficacy, such as the combined use of different ICIs, are being tested in many ongoing clinical trials. Repertoire analysis is expected to be performed in these trials. It is also important to study the effects of tumor heterogeneity and symbiotic bacteria (e.g., intestinal bacteria) on immune cell repertoires. Further studies are necessary to determine whether the results of immune cell repertoire analysis in melanoma can be extrapolated to other types of cancers.

Author Contributions

K.N. and R.O. have equally contributed to the writing and editing of this manuscript. All authors have read and agreed to the published version of the manuscript.

Funding

This research received no external funding.

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Ribas, A.; Wolchok, J.D. Cancer immunotherapy using checkpoint blockade. Science 2018, 359, 1350–1355. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  2. Blank, C.U.; Haining, W.N.; Held, W.; Hogan, P.G.; Kallies, A.; Lugli, E.; Lynn, R.C.; Philip, M.; Rao, A.; Restifo, N.P.; et al. Defining ‘T cell exhaustion’. Nat. Rev. Immunol. 2019, 19, 665–674. [Google Scholar] [CrossRef] [PubMed]
  3. Carretero-González, A.; Lora, D.; Ghanem, I.; Zugazagoitia, J.; Castellano, D.; Sepúlveda, J.M.; López-Martin, J.A.; Paz-Ares, L.; de Velasco, G. Analysis of response rate with ANTI PD1/PD-L1 monoclonal antibodies in advanced solid tumors: A meta-analysis of randomized clinical trials. Oncotarget 2018, 9, 8706–8715. [Google Scholar] [CrossRef] [Green Version]
  4. Van der Leun, A.M.; Thommen, D.S.; Schumacher, T.N. CD8+ T cell states in human cancer: Insights from single-cell analysis. Nat. Rev. Cancer 2020, 20, 218–232. [Google Scholar] [CrossRef] [PubMed]
  5. Barnes, T.A.; Amir, E. HYPE or HOPE: The prognostic value of infiltrating immune cells in cancer. Br. J. Cancer 2017, 117, 451–460. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  6. Kirsch, I.; Vignali, M.; Robins, H. T-cell receptor profiling in cancer. Mol. Oncol. 2015, 9, 2063–2070. [Google Scholar] [CrossRef] [PubMed]
  7. Valpione, S.; Galvani, E.; Tweedy, J.; Mundra, P.A.; Banyard, A.; Middlehurst, P.; Barry, J.; Mills, S.; Salih, Z.; Weightman, J.; et al. Immune- awakening revealed by peripheral T cell dynamics after one cycle of immunotherapy. Nat. Cancer 2020, 1, 210–221. [Google Scholar] [CrossRef] [PubMed]
  8. Yost, K.E.; Satpathy, A.T.; Wells, D.K.; Qi, Y.; Wang, C.; Kageyama, R.; McNamara, K.L.; Granja, J.M.; Sarin, K.Y.; Brown, R.A.; et al. Clonal replacement of tumor-specific T cells following PD-1 blockade. Nat. Med. 2019, 25, 1251–1259. [Google Scholar] [CrossRef] [PubMed]
  9. Huang, A.C.; Postow, M.A.; Orlowski, R.J.; Mick, R.; Bengsch, B.; Manne, S.; Xu, W.; Harmon, S.; Giles, J.R.; Wenz, B.; et al. T-cell invigoration to tumour burden ratio associated with anti-PD-1 response. Nature 2017, 545, 60–65. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  10. Lythe, G.; Callard, R.E.; Hoare, R.L.; Molina-París, C. How many TCR clonotypes does a body maintain? J. Theor. Biol. 2016, 389, 214–224. [Google Scholar] [CrossRef]
  11. Kappler, J.; Kubo, R.; Haskins, K.; Hannum, C.; Marrack, P.; Pigeon, M.; McIntyre, B.; Allison, J.; Trowbridge, I. The major histocompatibility complex-restricted antigen receptor on T cells in mouse and man: Identification of constant and variable peptides. Cell 1983, 35, 295–302. [Google Scholar] [CrossRef]
  12. Monks, C.R.; Freiberg, B.A.; Kupfer, H.; Sciaky, N.; Kupfer, A. Three-dimensional segregation of supramolecular activation clusters in T cells. Nature 1998, 395, 82–86. [Google Scholar] [CrossRef]
  13. Boon, T.; van der Bruggen, P. Human tumor antigens recognized by T lymphocytes. J. Exp. Med. 1996, 183, 725–729. [Google Scholar] [CrossRef] [PubMed]
  14. Gudmundsdottir, H.; Wells, A.D.; Turka, L.A. Dynamics and requirements of T cell clonal expansion in vivo at the single-cell level: Effector function is linked to proliferative capacity. J. Immunol. 1999, 162, 5212–5223. [Google Scholar]
  15. Rizvi, N.A.; Hellmann, M.D.; Snyder, A.; Kvistborg, P.; Makarov, V.; Havel, J.J.; Lee, W.; Yuan, J.; Wong, P.; Ho, T.S.; et al. Cancer immunology. Mutational landscape determines sensitivity to PD-1 blockade in non-small cell lung cancer. Science 2015, 348, 124–128. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  16. Hozumi, N.; Tonegawa, S. Evidence for somatic rearrangement of immunoglobulin genes coding for variable and constant regions. Proc. Natl. Acad. Sci. USA 1976, 73, 3628–3632. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  17. Schatz, D.G.; Baltimore, D. Uncovering the V(D)J recombinase. Cell 2004, 2, S103–S108. [Google Scholar] [CrossRef] [Green Version]
  18. Boon, T.; Cerottini, J.C.; Van den Eynde, B.; van der Bruggen, P.; Van Pel, A. Tumor antigens recognized by T lymphocytes. Annu. Rev. Immunol. 1994, 12, 337–365. [Google Scholar] [CrossRef] [PubMed]
  19. Gros, A.; Parkhurst, M.R.; Tran, E.; Pasetto, A.; Robbins, P.F.; Ilyas, S.; Prickett, T.D.; Gartner, J.J.; Crystal, J.S.; Roberts, I.M.; et al. Prospective identification of neoantigen-specific lymphocytes in the peripheral blood of melanoma patients. Nat. Med. 2016, 22, 433–438. [Google Scholar] [CrossRef] [PubMed]
  20. Li, T.; Zhao, L.; Yang, Y.; Wang, Y.; Zhang, Y.; Guo, J.; Chen, G.; Qin, P.; Xu, B.; Ma, B.; et al. T cells expanded from PD-1+ peripheral blood lymphocytes share more clones with paired tumor-infiltrating lymphocytes. Cancer Res. 2021, 81, 2184–2194. [Google Scholar] [CrossRef]
  21. Arakawa, A.; Vollmer, S.; Tietze, J.; Galinski, A.; Heppt, M.V.; Bürdek, M.; Berking, C.; Prinz, J.C. Clonality of CD4+ Blood T Cells Predicts Longer Survival With CTLA4 or PD-1 Checkpoint Inhibition in Advanced Melanoma. Front. Immunol. 2019, 10, 1336. [Google Scholar] [CrossRef] [PubMed]
  22. Lucca, L.E.; Axisa, P.P.; Lu, B.; Harnett, B.; Jessel, S.; Zhang, L.; Raddassi, K.; Zhang, L.; Olino, K.; Clune, J.; et al. Circulating clonally expanded T cells reflect functions of tumor-infiltrating T cells. J. Exp. Med. 2021, 218, e20200921. [Google Scholar] [CrossRef] [PubMed]
  23. Magnuson, A.M.; Kiner, E.; Ergun, A.; Park, J.S.; Asinovski, N.; Ortiz-Lopez, A.; Kilcoyne, A.; Paoluzzi-Tomada, E.; Weissleder, R.; Mathis, D.; et al. Identification and validation of a tumor-infiltrating Treg transcriptional signature conserved across species and tumor types. Proc. Natl. Acad. Sci. USA 2018, 115, E10672–E10681. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  24. Kim, H.J.; Cantor, H. CD4 T-cell subsets and tumor immunity: The helpful and the not-so-helpful. Cancer Immunol. Res. 2014, 2, 91–98. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  25. Tay, R.E.; Richardson, E.K.; Toh, H.C. Revisiting the Role of CD4+ T Cells in Cancer Immunotherapy—New Insights into Old Paradigms. Cancer Gene Ther. 2021, 28, 5–17. [Google Scholar] [CrossRef]
  26. Sautès-Fridman, C.; Petitprez, F.; Calderaro, J.; Fridman, W.H. Tertiary lymphoid structures in the era of cancer immunotherapy. Nat. Rev. Cancer 2019, 19, 307–325. [Google Scholar] [CrossRef] [PubMed]
  27. Baecher-Allan, C.D.; Viglietta, V.; Hafler, D.A.; Human, C.D. Human CD4+CD25+ regulatory T cells. Semin. Immunol. 2004, 16, 89–98. [Google Scholar] [CrossRef]
  28. Sakaguchi, S.; Yamaguchi, T.; Nomura, T.; Ono, M. Regulatory T cells and immune tolerance. Cell 2008, 133, 775–787. [Google Scholar] [CrossRef] [Green Version]
  29. Ahmadzadeh, M.; Felipe-Silva, A.; Heemskerk, B.; Powell, D.J.; Wunderlich, J.R.; Merino, M.J.; Rosenberg, S.A. FOXP3 expression accurately defines the population of intratumoral regulatory T cells that selectively accumulate in metastatic melanoma lesions. Blood 2008, 112, 4953–4960. [Google Scholar] [CrossRef] [PubMed]
  30. Ahmadzadeh, M.; Pasetto, A.; Jia, L.; Deniger, D.C.; Stevanović, S.; Robbins, P.F.; Rosenberg, S.A. Tumor-infiltrating human CD4+ regulatory T cells display a distinct TCR repertoire and exhibit tumor and neoantigen reactivity. Sci. Immunol. 2019, 4, eaao4310. [Google Scholar] [CrossRef] [PubMed]
  31. Im, S.J.; Hashimoto, M.; Gerner, M.Y.; Lee, J.; Kissick, H.T.; Burger, M.C.; Shan, Q.; Hale, J.S.; Lee, J.; Nasti, T.H.; et al. Defining CD8+ T cells that provide the proliferative burst after PD-1 therapy. Nature 2016, 537, 417–421. [Google Scholar] [CrossRef]
  32. Mandric, I.; Rotman, J.; Yang, H.T.; Strauli, N.; Montoya, D.J.; Van Der Wey, W.; Ronas, J.R.; Statz, B.; Yao, D.; Petrova, V.; et al. Profiling immunoglobulin repertoires across multiple human tissues using RNA sequencing. Nat. Commun. 2020, 11, 3126. [Google Scholar] [CrossRef] [PubMed]
  33. Sade-Feldman, M.; Yizhak, K.; Bjorgaard, S.L.; Ray, J.P.; de Boer, C.G.; Jenkins, R.W.; Lieb, D.J.; Chen, J.H.; Frederick, D.T.; Barzily-Rokni, M.; et al. Defining T cell states associated with response to checkpoint immunotherapy in melanoma. Cell 2018, 175, 998–1013.e20. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  34. Peiffer, L.; Farahpour, F.; Sriram, A.; Spassova, I.; Hoffmann, D.; Kubat, L.; Stoitzner, P.; Gambichler, T.; Sucker, A.; Ugurel, S.; et al. BRAF and MEK inhibition in melanoma patients enables reprogramming of tumor infiltrating lymphocytes. Cancer Immunol. Immunother. 2021, 70, 1635–1647. [Google Scholar] [CrossRef] [PubMed]
  35. Jeannet, G.; Boudousquié, C.; Gardiol, N.; Kang, J.; Huelsken, J.; Held, W. Essential role of the Wnt pathway effector Tcf-1 for the establishment of functional CD8 T cell Memory. Proc. Natl. Acad. Sci. USA 2010, 107, 9777–9782. [Google Scholar] [CrossRef] [Green Version]
  36. Sullivan, B.M.; Juedes, A.; Szabo, S.J.; von Herrath, M.; Glimcher, L.H. Antigen-driven effector CD8 T cell function regulated by T-bet. Proc. Natl. Acad. Sci. USA 2003, 100, 15818–15823. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  37. Schenkel, J.M.; Masopust, D.; Memory, T.-R. Tissue-resident memory T cells. Immunity 2014, 41, 886–897. [Google Scholar] [CrossRef] [Green Version]
  38. Park, C.O.; Kupper, T.S. The emerging role of resident Memory T cells in protective immunity and inflammatory disease. Nat. Med. 2015, 21, 688–697. [Google Scholar] [CrossRef] [Green Version]
  39. Edwards, J.; Wilmott, J.S.; Madore, J.; Gide, T.N.; Quek, C.; Tasker, A.; Ferguson, A.; Chen, J.; Hewavisenti, R.; Hersey, P.; et al. CD103+ tumor-resident CD8+ T cells are associated with improved survival in immunotherapy-naïve melanoma patients and expand significantly during anti-PD-1 treatment. Clin. Cancer Res. 2018, 24, 3036–3045. [Google Scholar] [CrossRef] [Green Version]
  40. Smazynski, J.; Webb, J.R. Resident Memory-Like Tumor-Infiltrating Lymphocytes (TILRM): Latest players in the immuno-oncology repertoire. Front. Immunol. 2018, 9, 1741. [Google Scholar] [CrossRef] [Green Version]
  41. Boddupalli, C.S.; Bar, N.; Kadaveru, K.; Krauthammer, M.; Pornputtapong, N.; Mai, Z.; Ariyan, S.; Narayan, D.; Kluger, H.; Deng, Y.; 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. [Google Scholar] [CrossRef] [PubMed]
  42. Wu, T.D.; Madireddi, S.; de Almeida, P.E.; Banchereau, R.; Chen, Y.J.; Chitre, A.S.; Chiang, E.Y.; Iftikhar, H.; O’Gorman, W.E.; Au-Yeung, A.; et al. Peripheral T cell expansion predicts tumour infiltration and clinical response. Nature 2020, 579, 274–278. [Google Scholar] [CrossRef] [PubMed]
  43. Han, J.; Zhao, Y.; Shirai, K.; Molodtsov, A.; Kolling, F.W.; Fisher, J.L.; Zhang, P.; Yan, S.; Searles, T.G.; Bader, J.M.; et al. Resident and circulating memory T cells persist for years in melanoma patients with durable responses to immunotherapy. Nat. Cancer 2021, 2, 300–311. [Google Scholar] [CrossRef] [PubMed]
  44. Kurosaki, T. Genetic analysis of B cell antigen receptor signaling. Annu. Rev. Immunol. 1999, 17, 555–592. [Google Scholar] [CrossRef] [PubMed]
  45. Somasundaram, R.; Zhang, G.; Fukunaga-Kalabis, M.; Perego, M.; Krepler, C.; Xu, X.; Wagner, C.; Hristova, D.; Zhang, J.; Tian, T.; et al. Tumor-associated B-cells induce tumor heterogeneity and therapy resistance. Nat. Commun. 2017, 8, 607. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  46. Griss, J.; Bauer, W.; Wagner, C.; Simon, M.; Chen, M.; Grabmeier-Pfistershammer, K.; Maurer-Granofszky, M.; Roka, F.; Penz, T.; Bock, C.; et al. B cells sustain inflammation and predict response to immune checkpoint blockade in human melanoma. Nat. Commun. 2019, 10, 4186. [Google Scholar] [CrossRef] [Green Version]
  47. Cabrita, R.; Lauss, M.; Sanna, A.; Donia, M.; Skaarup Larsen, M.; Mitra, S.; Johansson, I.; Phung, B.; Harbst, K.; Vallon-Christersson, J.; et al. Tertiary lymphoid structures improve immunotherapy and survival in melanoma. Nature 2020, 577, 561–565. [Google Scholar] [CrossRef]
  48. Helmink, B.A.; Reddy, S.M.; Gao, J.; Zhang, S.; Basar, R.; Thakur, R.; Yizhak, K.; Sade-Feldman, M.; Blando, J.; Han, G.; et al. B cells and tertiary lymphoid structures promote immunotherapy response. Nature 2020, 577, 549–555. [Google Scholar] [CrossRef]
  49. Anagnostou, V.; Bruhm, D.C.; Niknafs, N.; White, J.R.; Shao, X.M.; Sidhom, J.W.; Stein, J.; Tsai, H.L.; Wang, H.; Belcaid, Z.; et al. Integrative tumor and immune cell multi-omic analyses predict response to immune checkpoint blockade in melanoma. Cell Reprod. Med. 2020, 1, 100139. [Google Scholar] [CrossRef]
  50. Lee, J.; Ahn, E.; Kissick, H.T.; Ahmed, R. Reinvigorating exhausted T cells by blockade of the PD-1 pathway. For. Immunopathol. Dis. Therap. 2015, 6, 7–17. [Google Scholar] [CrossRef] [Green Version]
  51. Francisco, L.M.; Salinas, V.H.; Brown, K.E.; Vanguri, V.K.; Freeman, G.J.; Kuchroo, V.K.; Sharpe, A.H. PD-L1 regulates the development, maintenance, and function of induced regulatory T cells. J. Exp. Med. 2009, 206, 3015–3029. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  52. Nguyen, L.T.; Ohashi, P.S. Clinical blockade of PD1 and LAG3-potential mechanisms of action. Nat. Rev. Immunol. 2015, 15, 45–56. [Google Scholar] [CrossRef] [PubMed]
  53. Messal, N.; Serriari, N.E.; Pastor, S.; Nunès, J.A.; Olive, D. PD-L2 is expressed on activated human T cells and regulates their function. Mol. Immunol. 2011, 48, 2214–2219. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  54. Inoue, H.; Park, J.H.; Kiyotani, K.; Zewde, M.; Miyashita, A.; Jinnin, M.; Kiniwa, Y.; Okuyama, R.; Tanaka, R.; Fujisawa, Y.; et al. Intratumoral expression levels of PD-L1, GZMA, and HLA-A along with oligoclonal T cell expansion associate with response to nivolumab in metastatic melanoma. Oncoimmunology 2016, 30, e1204507. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  55. Huang, A.C.; Orlowski, R.J.; Xu, X.; Mick, R.; George, S.M.; Yan, P.K.; Manne, S.; Kraya, A.A.; Wubbenhorst, B.; Dorfman, L.; et al. A single dose of neoadjuvant PD-1 blockade predicts clinical outcomes in resectable melanoma. Nat. Med. 2019, 25, 454–461. [Google Scholar] [CrossRef] [PubMed]
  56. Gajewski, T.F.; Fallarino, F.; Fields, P.E.; Rivas, F.; Alegre, M.L. Absence of CTLA-4 lowers the activation threshold of primed CD8þ TCR-transgenic T cells: Lack of correlation with Src homology domain 2-containing protein tyrosine phosphatase. J. Immunol. 2001, 166, 3900–3907. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  57. Pedicord, V.A.; Montalvo, W.; Leiner, I.M.; Allison, J.P. Single dose of anti-CTLA-4 enhances CD8þ T-cell memory formation, function, and maintenance. Proc. Natl. Acad. Sci. USA 2011, 108, 266–271. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  58. Read, S.; Greenwald, R.; Izcue, A.; Robinson, N.; Mandelbrot, D.; Francisco, L.; Sharpe, A.H.; Powrie, F. Blockade of CTLA-4 on CD4þCD25þ regulatory T cells abrogates their function in vivo. J. Immunol. 2006, 177, 4376–4383. [Google Scholar] [CrossRef] [Green Version]
  59. Noyes, D.; Bag, A.; Oseni, S.; Semidey-Hurtado, J.; Cen, L.; Sarnaik, A.A.; Sondak, V.K.; Adeegbe, D. Tumor-associated Tregs obstruct antitumor immunity by promoting T cell dysfunction and restricting clonal diversity in tumor-infiltrating CD8+ T cells. J. Immunother. Cancer 2022, 10, e004605. [Google Scholar] [CrossRef]
  60. Twyman-Saint Victor, C.; Rech, A.J.; Maity, A.; Rengan, R.; Pauken, K.E.; Stelekati, E.; Benci, J.L.; Xu, B.; Dada, H.; Odorizzi, P.M.; et al. Radiation and dual checkpoint blockade activate non-redundant immune mechanisms in cancer. Nature 2015, 520, 373–377. [Google Scholar] [CrossRef] [Green Version]
  61. Cha, E.; Klinger, M.; Hou, Y.; Cummings, C.; Ribas, A.; Faham, M.; Fong, L. Improved survival with T cell clonotype stability after anti-CTLA-4 treatment in cancer patients. Sci. Transl. Med. 2014, 6, 238ra70. [Google Scholar] [CrossRef] [PubMed]
  62. Robert, L.; Tsoi, J.; Wang, X.; Emerson, R.; Homet, B.; Chodon, T.; Mok, S.; Huang, R.R.; Cochran, A.J.; Comin-Anduix, B.; et al. CTLA4 Blockade Broadens the Peripheral T Cell Receptor Repertoire. Clin. Cancer Res. 2014, 20, 2424–2432. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  63. Topalian, S.L.; Hodi, F.S.; Brahmer, J.R.; Gettinger, S.N.; Smith, D.C.; McDermott, D.F.; Powderly, J.D.; Carvajal, R.D.; Sosman, J.A.; Atkins, M.B.; et al. Safety, activity, and immune correlates of anti-PD-1 antibody in cancer. N. Engl. J. Med. 2012, 366, 2443–2454. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  64. Anagnostou, V.; Smith, K.N.; Forde, P.M.; Niknafs, N.; Bhattacharya, R.; White, J.; Zhang, T.; Adleff, V.; Phallen, J.; Wali, N.; et al. Evolution of neoantigen landscape during immune checkpoint blockade in non-small cell lung cancer. Cancer Discov. 2017, 7, 264–276. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  65. Nagasaki, J.; Inozume, T.; Sax, N.; Ariyasu, R.; Ishikawa, M.; Yamashita, K.; Kawazu, M.; Ueno, T.; Irie, T.; Tanji, E.; et al. PD-1 blockade therapy promotes infiltration of tumor-attacking exhausted T cell clonotypes. Cell Rep. 2022, 38, 110331. [Google Scholar] [CrossRef] [PubMed]
  66. D’Souza, W.N.; Chang, C.F.; Fischer, A.M.; Li, M.; Hedrick, S.M. The Erk2 MAPK regulates CD8 T cell proliferation and survival. J. Immunol. 2008, 181, 7617–7629. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  67. Dushyanthen, S.; Teo, Z.L.; Caramia, F.; Savas, P.; Mintoff, C.P.; Virassamy, B.; Henderson, M.A.; Luen, S.J.; Mansour, M.; Kershaw, M.H.; et al. Agonist immunotherapy restores T cell function following MEK inhibition improving efficacy in breast cancer. Nat. Commun. 2017, 8, 606. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  68. Hu-Lieskovan, S.; Mok, S.; Homet Moreno, B.; Tsoi, J.; Robert, L.; Goedert, L.; Pinheiro, E.M.; Koya, R.C.; Graeber, T.G.; Comin-Anduix, B.; et al. Improved antitumor activity of immunotherapy with BRAF and MEK inhibitors in BRAF(V600E) melanoma. Sci. Transl. Med. 2015, 7, 279ra41. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  69. Ebert, P.J.R.; Cheung, J.; Yang, Y.; McNamara, E.; Hong, R.; Moskalenko, M.; Gould, S.E.; Maecker, H.; Irving, B.A.; Kim, J.M.; et al. MAP kinase inhibition promotes T cell and anti-tumor activity in combination with PD-L1 checkpoint blockade. Immunity 2016, 44, 609–621. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  70. Cooper, Z.A.; Frederick, D.T.; Juneja, V.R.; Sullivan, R.J.; Lawrence, D.P.; Piris, A.; Sharpe, A.H.; Fisher, D.E.; Flaherty, K.T.; Wargo, J.A. BRAF inhibition is associated with increased clonality in tumor-infiltrating lymphocytes. Oncoimmunology 2013, 2, e26615. [Google Scholar] [CrossRef] [Green Version]
  71. Seitter, S.J.; Sherry, R.M.; Yang, J.C.; Robbins, P.F.; Shindorf, M.L.; Copeland, A.R.; McGowan, C.T.; Epstein, M.; Shelton, T.E.; Langhan, M.M.; et al. Impact of Prior Treatment on the Efficacy of Adoptive Transfer of Tumor-Infiltrating Lymphocytes in Patients with Metastatic Melanoma. Clin. Cancer Res. 2021, 27, 5289–5298. [Google Scholar] [CrossRef]
  72. Robbins, P.F.; Lu, Y.C.; El-Gamil, M.; Li, Y.F.; Gross, C.; Gartner, J.; Lin, J.C.; Teer, J.K.; Cliften, P.; Tycksen, E.; et al. Mining exomic sequencing data to identify mutated antigens recognized by adoptively transferred tumor-reactive T cells. Nat. Med. 2013, 19, 747–752. [Google Scholar] [CrossRef]
  73. Lauss, M.; Donia, M.; Harbst, K.; Andersen, R.; Mitra, S.; Rosengren, F.; Salim, M.; Vallon-Christersson, J.; Torngren, T.; Kvist, A.; et al. Mutational and putative neoantigen load predict clinical benefit of adoptive T cell therapy in melanoma. Nat. Commun. 2017, 8, 1738. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  74. Levi, S.T.; Copeland, A.R.; Nah, S.; Crystal, J.S.; Ivey, G.D.; Lalani, A.; Jafferji, M.; White, B.S.; Parikh, N.B.; Leko, V.; et al. Neoantigen Identification and Response to Adoptive Cell Transfer in anti PD-1 Naïve and Experienced Patients with Metastatic Melanoma. Clin. Cancer Res. 2022, 28, 3042–3052. [Google Scholar] [CrossRef] [PubMed]
  75. Kobayashi, M.; Fitz, L.; Ryan, M.; Hewick, R.M.; Clark, S.C.; Chan, S.; Loudon, R.; Sherman, F.; Perussia, B.; Trinchieri, G. Identification and purification of natural killer cell stimulatory factor (NKSF), a cytokine with multiple biologic effects on human lymphocytes. J. Exp. Med. 1989, 170, 827–845. [Google Scholar] [CrossRef]
  76. Aste-Amezaga, M.; D’Andrea, A.; Kubin, M.; Trinchieri, G. Cooperation of natural killer cell stimulatory factor/interleukin-12 with other stimuli in the induction of cytokines and cytotoxic cell-associated molecules in human T and NK cells. Cell Immunol. 1994, 156, 480–492. [Google Scholar] [CrossRef]
  77. Chan, S.H.; Perussia, B.; Gupta, J.W.; Kobayashi, M.; Pospisil, M.; Young, H.A.; Wolf, S.F.; Young, D.; Clark, S.C.; Trinchieri, G. Induction of interferon gamma production by natural killer cell stimulatory factor: Characterization of the responder cells and synergy with other inducers. J. Exp. Med. 1991, 173, 869–879. [Google Scholar] [CrossRef] [Green Version]
  78. Hsieh, C.S.; Macatonia, S.E.; Tripp, C.S.; Wolf, S.F.; O’Garra, A.; Murphy, K.M. Development of TH1 CD4+ T cells through IL-12 produced by Listeria-induced macrophages. Science 1993, 260, 547–549. [Google Scholar] [CrossRef]
  79. Airoldi, I.; Di Carlo, E.; Cocco, C.; Sorrentino, C.; Fais, F.; Cilli, M.; D’Antuono, T.; Colombo, M.P.; Pistoia, V. Lack of Il12rb2 signaling predisposes to spontaneous autoimmunity and malignancy. Blood 2005, 106, 3846–3853. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  80. Greaney, S.K.; Algazi, A.P.; Tsai, K.K.; Takamura, K.T.; Chen, L.; Twitty, C.G.; Zhang, L.; Paciorek, A.; Pierce, R.H.; Le, M.H.; et al. Intratumoral Plasmid IL12 Electroporation Therapy in Patients with Advanced Melanoma Induces Systemic and Intratumoral T-cell Responses. Cancer Immunol. Res. 2020, 8, 246–254. [Google Scholar] [CrossRef] [Green Version]
  81. Chambers, C.A.; Kuhns, M.S.; Egen, J.G.; Allison, J.P. CTLA-4-mediated inhibition in regulation of T cell responses: Mechanisms and manipulation in tumor immunotherapy. Annu. Rev. Immunol. 2001, 19, 565–594. [Google Scholar] [CrossRef] [PubMed]
  82. Nishimura, H.; Nose, M.; Hiai, H.; Minato, N.; Honjo, T. Development of Lupus-like Autoimmune Diseases by Disruption of the PD-1 Gene Encoding an ITIM Motif-Carrying Immunoreceptor. Immunity 1999, 11, 141–151. [Google Scholar] [CrossRef] [Green Version]
  83. Freeman, G.J.; Long, A.J.; Iwai, Y.; Bourque, K.; Chernova, T.; Nishimura, H.; Fitz, L.J.; Malenkovich, N.; Okazaki, T.; Byrne, M.C.; et al. Engagement of the PD-1 immunoinhibitory receptor by a novel B7 family member leads to negative regulation of lymphocyte activation. J. Exp. Med. 2000, 192, 1027–1034. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  84. Robins, H.S.; Campregher, P.V.; Srivastava, S.K.; Wacher, A.; Turtle, C.J.; Kahsai, O.; Riddell, S.R.; Warren, E.H.; Carlson, C.S. Comprehensive assessment of T-cell receptor beta-chain diversity in alphabeta T cells. Blood 2009, 114, 4099–4107. [Google Scholar] [CrossRef] [PubMed]
  85. Valpione, S.; Mundra, P.A.; Galvani, E.; Campana, L.G.; Lorigan, P.; De Rosa, F.; Gupta, A.; Weightman, J.; Mills, S.; Dhomen, N.; et al. The T cell receptor repertoire of tumor infiltrating T cells is predictive and prognostic for cancer survival. Nat. Commun. 2021, 12, 4098. [Google Scholar] [CrossRef] [PubMed]
  86. Hogan, S.A.; Courtier, A.; Cheng, P.F.; Jaberg-Bentele, N.F.; Goldinger, S.M.; Manuel, M.; Perez, S.; Plantier, N.; Mouret, J.F.; Nguyen-Kim, T.D.L.; et al. Peripheral Blood TCR Repertoire Profiling May Facilitate Patient Stratification for Immunotherapy against Melanoma. Cancer Immunol. Res. 2019, 7, 77–85. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  87. Hodi, F.S.; O’Day, S.J.; McDermott, D.F.; Weber, R.W.; Sosman, J.A.; Haanen, J.B.; Gonzalez, R.; Robert, C.; Schadendorf, D.; Hassel, J.C.; et al. Improved survival with ipilimumab in patients with metastatic melanoma. N. Engl. J. Med. 2010, 363, 711–723. [Google Scholar] [CrossRef]
  88. Oh, D.Y.; Cham, J.; Zhang, L.; Fong, G.; Kwek, S.S.; Klinger, M.; Faham, M.; Fong, L. 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. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  89. Subudhi, S.K.; Aparicio, A.; Gao, J.; Zurita, A.J.; Araujo, J.C.; Logothetis, C.J.; Tahir, S.A.; Korivi, B.R.; Slack, R.S.; Vence, L.; et al. Clonal expansion of CD8 T cells in the systemic circulation precedes development of ipilimumab-induced toxicities. Proc. Natl. Acad. Sci. USA 2016, 113, 11919–11924. [Google Scholar] [CrossRef] [Green Version]
  90. Ott, P.A.; Hu, Z.; Keskin, D.B.; Shukla, S.A.; Sun, J.; Bozym, D.J.; Zhang, W.; Luoma, A.; Giobbie-Hurder, A.; Peter, L.; et al. An immunogenic personal neoantigen vaccine for patients with melanoma. Nature 2017, 547, 217–221. [Google Scholar] [CrossRef] [Green Version]
  91. Fritsch, E.F.; Rajasagi, M.; Ott, P.A.; Brusic, V.; Hacohen, N.; Wu, C.J. HLA- binding properties of tumor neoepitopes in humans. Cancer Immunol. Res. 2014, 2, 522–529. [Google Scholar] [CrossRef] [PubMed]
  92. Abelin, J.G.; Harjanto, D.; Malloy, M.; Suri, P.; Colson, T.; Goulding, S.P.; Creech, A.L.; Serrano, L.R.; Nasir, G.; Nasrullah, Y.; et al. Defining HLA- II. Immunity 2019, 51, 766–779.e17. [Google Scholar] [CrossRef] [PubMed]
  93. Racle, J.; Michaux, J.; Rockinger, G.A.; Arnaud, M.; Bobisse, S.; Chong, C.; Guillaume, P.; Coukos, G.; Harari, A.; Jandus, C.; et al. Robust Prediction of HLA class II Epitopes by Deep Motif Deconvolution of Immunopeptidomes. Nat. Biotechnol. 2019, 37, 1283–1286. [Google Scholar] [CrossRef] [PubMed]
  94. Reynisson, B.; Barra, C.; Kaabinejadian, S.; Hildebrand, W.H.; Peters, B.; Nielsen, M. Improved prediction of MHC II antigen presentation through integration and motif deconvolution of mass spectrometry MHC eluted ligand data. J. Proteome Res. 2020, 19, 2304–2315. [Google Scholar] [CrossRef] [PubMed]
  95. Riaz, N.; Havel, J.J.; Makarov, V.; Desrichard, A.; Urba, W.J.; Sims, J.S.; Hodi, F.S.; Martín-Algarra, S.; Mandal, R.; Sharfman, W.H.; et al. Tumor and Microenvironment Evolution during Immunotherapy with Nivolumab. Cell 2017, 171, 934–949.e16. [Google Scholar] [CrossRef] [Green Version]
  96. Yuzhakova, D.V.; Volchkova, L.N.; Pogorelyy, M.V.; Serebrovskaya, E.O.; Shagina, I.A.; Bryushkova, E.A.; Nakonechnaya, T.O.; Izosimova, A.V.; Zavyalova, D.S.; Karabut, M.M.; et al. Measuring Intratumoral Heterogeneity of Immune Repertoires. Front. Oncol. 2020, 10, 512. [Google Scholar] [CrossRef]
  97. Routy, B.; Le Chatelier, E.; Derosa, L.; Duong, C.P.M.; Alou, M.T.; Daillère, R.; Fluckiger, A.; Messaoudene, M.; Rauber, C.; Roberti, M.P.; et al. Gut microbiome influences efficacy of PD-1-based immunotherapy against epithelial tumors. Science 2018, 359, 91–97. [Google Scholar] [CrossRef] [Green Version]
  98. Gopalakrishnan, V.; Spencer, C.N.; Nezi, L.; Reuben, A.; Andrews, M.C.; Karpinets, T.V.; Prieto, P.A.; Vicente, D.; Hoffman, K.; Wei, S.C.; et al. Gut microbiome modulates response to anti-PD-1 immunotherapy in melanoma patients. Science 2018, 359, 97–103. [Google Scholar] [CrossRef] [Green Version]
  99. Iida, N.; Dzutsev, A.; Stewart, C.A.; Smith, L.; Bouladoux, N.; Weingarten, R.A.; Molina, D.A.; Salcedo, R.; Back, T.; Cramer, S.; et al. Commensal bacteria control cancer response to therapy by modulating the tumor microenvironment. Science 2013, 342, 967–970. [Google Scholar] [CrossRef]
  100. Bessell, C.A.; Isser, A.; Havel, J.J.; Lee, S.; Bell, D.R.; Hickey, J.W.; Chaisawangwong, W.; Glick Bieler, J.; Srivastava, R.; Kuo, F.; et al. Commensal bacteria stimulate antitumor responses via T cell cross-reactivity. JCI Insight 2020, 5, e135597. [Google Scholar] [CrossRef]
  101. Baruch, E.N.; Youngster, I.; Ben-Betzalel, G.; Ortenberg, R.; Lahat, A.; Katz, L.; Adler, K.; Dick-Necula, D.; Raskin, S.; Bloch, N.; et al. Fecal microbiota transplant promotes response in immunotherapy-refractory melanoma patients. Science 2021, 371, 602–609. [Google Scholar] [CrossRef] [PubMed]
  102. Nejman, D.; Livyatan, I.; Fuks, G.; Gavert, N.; Zwang, Y.; Geller, L.T.; Rotter-Maskowitz, A.; Weiser, R.; Mallel, G.; Gigi, E.; et al. The human tumor microbiome is composed of tumor type-specific intracellular bacteria. Science 2020, 368, 973–980. [Google Scholar] [CrossRef] [PubMed]
  103. Riquelme, E.; Zhang, Y.; Zhang, L.; Montiel, M.; Zoltan, M.; Dong, W.; Quesada, P.; Sahin, I.; Chandra, V.; San Lucas, A.; et al. Tumor microbiome diversity and composition influence pancreatic cancer outcomes. Cell 2019, 178, 795–806.e12. [Google Scholar] [CrossRef] [PubMed]
  104. Kalaora, S.; Nagler, A.; Nejman, D.; Alon, M.; Barbolin, C.; Barnea, E.; Ketelaars, S.L.C.; Cheng, K.; Vervier, K.; Shental, N.; et al. Identification of bacteria-derived HLA-bound peptides in melanoma. Nature 2021, 592, 138–143. [Google Scholar] [CrossRef] [PubMed]
  105. Alspach, E.; Lussier, D.M.; Miceli, A.P.; Kizhvatov, I.; DuPage, M.; Luoma, A.M.; Meng, W.; Lichti, C.F.; Esaulova, E.; Vomund, A.N.; et al. MHC-II neoantigens shape tumour immunity and response to immunotherapy. Nature 2019, 574, 696–701. [Google Scholar] [CrossRef]
  106. Rosato, P.C.; Wijeyesinghe, S.; Stolley, J.M.; Nelson, C.E.; Davis, R.L.; Manlove, L.S.; Pennell, C.A.; Blazar, B.R.; Chen, C.C.; Geller, M.A.; et al. Virus-specific memory T cells populate tumors and can be repurposed for tumor immunotherapy. Nat. Commun. 2019, 10, 567. [Google Scholar] [CrossRef] [Green Version]
  107. Chiaro, J.; Kasanen, H.H.E.; Whalley, T.; Capasso, C.; Grönholm, M.; Feola, S.; Peltonen, K.; Hamdan, F.; Hernberg, M.; Mäkelä, S.; et al. Viral Molecular Mimicry Influences the Antitumor Immune Response in Murine and Human Melanoma. Cancer Immunol. Res. 2021, 9, 981–993. [Google Scholar] [CrossRef]
  108. Oliveira, G.; Stromhaug, K.; Klaeger, S.; Kula, T.; Frederick, D.T.; Le, P.M.; Forman, J.; Huang, T.; Li, S.; Zhang, W.; et al. Phenotype, specificity and avidity of antitumour CD8+T cells in melanoma. Nature 2021, 596, 119–125. [Google Scholar] [CrossRef]
Figure 1. Repertoire changes with immune checkpoint inhibitors.
Figure 1. Repertoire changes with immune checkpoint inhibitors.
Ijms 23 12991 g001
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Share and Cite

MDPI and ACS Style

Nakamura, K.; Okuyama, R. Changes in the Immune Cell Repertoire for the Treatment of Malignant Melanoma. Int. J. Mol. Sci. 2022, 23, 12991. https://doi.org/10.3390/ijms232112991

AMA Style

Nakamura K, Okuyama R. Changes in the Immune Cell Repertoire for the Treatment of Malignant Melanoma. International Journal of Molecular Sciences. 2022; 23(21):12991. https://doi.org/10.3390/ijms232112991

Chicago/Turabian Style

Nakamura, Kenta, and Ryuhei Okuyama. 2022. "Changes in the Immune Cell Repertoire for the Treatment of Malignant Melanoma" International Journal of Molecular Sciences 23, no. 21: 12991. https://doi.org/10.3390/ijms232112991

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