Next Article in Journal
Inherited Eye Diseases with Retinal Manifestations through the Eyes of Homeobox Genes
Next Article in Special Issue
Abrogation of IFN-γ Signaling May not Worsen Sensitivity to PD-1/PD-L1 Blockade
Previous Article in Journal
Investigation of a Novel Salt Stress-Responsive Pathway Mediated by Arabidopsis DEAD-Box RNA Helicase Gene AtRH17 Using RNA-Seq Analysis
Previous Article in Special Issue
The Role of Immune Checkpoint Blockade in Uveal Melanoma
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Determinants of Resistance to Checkpoint Inhibitors

1
Department of Surgery (Urology), Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA
2
Cedars-Sinai Samuel Oschin Comprehensive Cancer Institute, Los Angeles, CA 90048, USA
3
Department of Pathology and Laboratory Medicine, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA
4
Cedars-Sinai Health System, 8700 Beverly Blvd., OCC Mezz C2002, Los Angeles, CA 90048, USA
*
Author to whom correspondence should be addressed.
Int. J. Mol. Sci. 2020, 21(5), 1594; https://doi.org/10.3390/ijms21051594
Submission received: 10 January 2020 / Revised: 22 February 2020 / Accepted: 23 February 2020 / Published: 26 February 2020
(This article belongs to the Special Issue Cancer Immunotherapy Using Checkpoint Inhibitors: Future Directions)

Abstract

:
The development of immune checkpoint inhibitors (ICIs) has drastically altered the landscape of cancer treatment. Since approval of the first ICI for the treatment of advanced melanoma in 2011, several therapeutic agents have been Food and Drug Administration (FDA)-approved for multiple cancers, and hundreds of clinical trials are currently ongoing. These antibodies disrupt T-cell inhibitory pathways established by tumor cells and thus re-activate the host’s antitumor immune response. While successful in many cancers, several types remain relatively refractory to treatment or patients develop early recurrence. Hence, there is a great need to further elucidate mechanisms of resistant disease and determine novel, effective, and tolerable combination therapies to enhance efficacy of ICIs.

1. Introduction

Because of their effectiveness, several immune checkpoint inhibitors (ICIs) are Food and Drug Administration (FDA)-approved for a wide range of cancer types [1]. Ipilimumab was the first ICI to be approved in 2011 for the treatment of advanced melanoma. This monoclonal antibody targets cytotoxic T-lymphocyte-associated protein 4 (CTLA-4), an inhibitory checkpoint on activated T cells. Similarly, monoclonal antibodies targeting the programmed cell death protein 1/programmed death-ligand 1 (PD-1/PD-L1) axis exploit a negative co-stimulatory signal meant to mitigate T-cell receptor (TCR) signaling. Blockade of these inhibitory signals reactivates the host antitumor immune response with consequent destruction of the tumor. The development of ICIs has drastically altered the landscape of cancer treatment and are especially appealing due to possible long-term durable responses. However, current therapies are not successful in all patients; even in generally responsive tumors and in certain tumor types, most patients do not respond or acquire early resistance. To increase efficacy, ICIs are being tested in combination with each other as well as other therapies targeting T-cell activation or pathways in the tumor microenvironment. In this review, we outline currently understood mechanisms of immune checkpoint inhibitor resistance and discuss the approaches used for discovery of rational combinations that can overcome such resistance.

2. Immune Checkpoint Inhibitors

2.1. CTLA-4

The human immune system has evolved pattern recognition receptors over a millennium to protect the host from foreign pathogens and developed mechanisms to screen immune cells from recognizing self-molecules. Adaptive cell-mediated immune responses are carried out by T cells, which are a type of lymphocyte that becomes activated to specific antigens to either directly (through cytotoxic CD8+ T cells) or indirectly (through helper CD4+ T cells) destroy abnormal cells. Overall, T helper (Th) cells regulate the activity of other immune cells by releasing T-cell cytokines. They are also important for B cell antibody class switching, activation, and growth of cytotoxic T cells, and in maximizing bactericidal activity of phagocytes, such as macrophages [2]. The cytokine secretion patterns of Th cells can further categorize these effector cells as type 1 (Th1) or type 2 (Th2), with type 1 producing interferon gamma (IFN-γ) and type 2 producing interleukin-4 (IL-4), IL-5, and IL-13 [3]. The activation of T cells requires engagement of a co-stimulatory molecule on a professional antigen presenting cell (APC) in addition to the TCR. Physiologically, this secondary signal prevents recognition of self-peptides, and the absence of such a signal results in T-cell anergy. Naïve T cells constitutively express CD28, the receptor for CD80 (B7-1) and CD86 (B7-2) proteins. Interaction of T-cell CD28 with B7-1 or B7-2 on APCs facilitates maturation, proliferation, activation, and survival of the former. Upon activation, T cells induce expression of CTLA-4 that attenuates signaling by competitively binding to B7-1 and B7-2 [4,5], with higher affinity than CD28; thus, inhibiting further activation and leading to immunological tolerance and prevention of autoimmunity. Following TCR signaling, additional CTLA-4 is recruited to the immunologic synapse via intracellular vesicles to spatially regulate inhibition [6]. CTLA-4 can also deplete available ligand from neighboring cells through trans-endocytosis. For example, B7-1 and B7-2 are degraded in CTLA-4+ cells, resulting in reduced co-stimulation through CD28 [7]. Immunotherapy using CTLA-4 blockade aims to sterically hinder interactions with B7 ligands [8] and thereby facilitate continued positive co-stimulation with CD28. This promotes antitumor response through expansion of tumor-infiltrating T cells and possibly the depletion of regulatory T-cell (Treg) populations, which are immunosuppressive and downregulate the activation and proliferation of effector T cells [9].

2.2. PD-1/PD-L1

PD-1 is a cell surface receptor expressed on activated T and B lymphocytes to regulate peripheral tolerance and T-cell responses. Binding of the receptor to its ligands PD-L1 or PD-L2 results in apoptosis of antigen-specific T cells, reduced TCR-mediated activation, attenuation of T-cell proliferation, and reduced apoptosis of Tregs, culminating in the inhibition of T-cell activity. These ligands are expressed on APCs and non-lymphoid tissues and can be induced by inflammatory cytokines, such as IFN-γ and IL-10 [10]. PD-L1 is frequently expressed on tumor cells, or in the tumor microenvironment, and allows evasion of immune detection [11]. The cytoplasmic region of PD-1 contains two tyrosine motifs, immunoreceptor tyrosine-based inhibition motif (ITIM) and immunoreceptor tyrosine-based switch motif (ITSM), which are phosphorylated upon receptor binding. Src homology 2-domain-containing tyrosine phosphatase 2 (SHP2) and SHP1 are then recruited to these phosphorylated motifs [12,13]. In T cells, PD-1 signaling results in dephosphorylation of key TCR signaling components, most notably CD28 and the ZAP70/CD3zeta signalosome [14,15]. This interference leads to reduced cytokine production, proliferation, and effector function. PD-1 blockade releases inhibition of TCR signaling and restores effector function of exhausted CD8+ T cells. The concept of “T-cell exhaustion”, first described during viral infections, is characterized by the stepwise and progressive loss of T-cell functions, and can culminate in the physical deletion of the responding cells. In cancer, these cells can remain in the microenvironment and exhibit reduced cytotoxic activity, disrupted cytokine production, and expression of inhibitory receptors [16]. Thus, PD-1 blockade supports a more effective antitumor immune response through reactivation of effector function. However, it is important to note that only certain subsets of exhausted T cells respond to PD-1/PD-L1 blockade, independent of PD-1 expression [15].

2.3. Additional T Cell Inhibitory and Stimulatory Pathways

While CTLA-4 and PD-1/PD-L1 ICIs have been shown to be effective as mono- and dual-therapeutic agents, other T-cell inhibitory and stimulatory pathways are currently being investigated for synergistic effect. Markers previously utilized to identify T-cell exhaustion are now being explored as potential targets for immune cell reactivation. Lymphocyte activation gene 3 (LAG-3) is a cell surface receptor expressed on activated T cells, B cells, natural killer cells, Tregs, and dendritic cells. LAG3 has structural homology to CD4 and can bind their shared ligand, major histocompatibility complex class II (MHC class-II) molecules, on APCs with a higher affinity [17]. LAG3 maintains T-cell homeostasis and negatively regulates their expansion [18]. T-cell immunoglobulin-3 (TIM-3) is a cell surface receptor expressed on activated T cells, Tregs, and white blood cells that mediate innate (non-specific) immunity. Engagement of TIM-3 in effector Th1 cells results in an influx of calcium and apoptosis [19]. T cell immunoglobulin and immunoreceptor tyrosine-based inhibitory motif domain (TIGIT) is a surface protein expressed on T cells and natural killer (NK) cells. Binding of TIGIT to poliovirus receptor inhibits natural killer cell cytotoxicity and inhibits T-cell activation through the maturation of immunoregulatory dendritic cells [20,21]. V-domain Ig suppressor of T-cell activation (VISTA) is an immune checkpoint molecule expressed on myeloid cells, T cells, and regulatory T cells. VISTA can function as an inhibitory receptor on T cells to suppress activation or as a ligand on APCs to modulate T-cell proliferation and cytokine production. Blockade of LAG-3, TIM-3, TIGIT, and VISTA have shown improved anti-tumor responses in preclinical models when combined with anti-CTLA-4 or anti-PD-1 therapy and are currently being evaluated in clinical trials. Detailed information on trials can be found at the https://clinicaltrials.gov/ web site.
Alternatively, agonists of T-cell activation are being explored to revitalize anticancer immune response. Several members of the tumor necrosis factor receptor superfamily (TNFRSF) have been identified as potential targets due to their functions in the regulation of immune cell populations implicated in cancer progression. TNFRSF4, or OX40, is expressed on CD4+ and CD8+ T cells, NK cells, and neutrophils. Stimulation of OX40 prolongs T-cell division and prevents apoptosis, resulting in increased numbers of surviving T cells that develop into memory T cells [22]. TNFRSF18, or glucocorticoid-induced TNF receptor-related protein (GITR), has been described in multiple immune cell types, most notably Tregs, and is found highly expressed on CD8+ and CD4+ T cells following TCR activation [23,24,25]. Signaling through GITR drives a high-avidity CD8+ T-cell response to tumor antigens, increases effector T-cell proliferation via expression of cytokines IL-2 and IFN-γ, and reduces the suppressive function of Tregs [26,27,28]. TNFRSF9, also referred to as 4-1BB or CD137, is expressed on NK cells, macrophages, B cells, and activated T cells. Stimulation of 4-1BB preferentially modulates CD8+ T cells, resulting in the amplification of populations in vivo and protection from activation-induced cell death [29,30]. TNFRSF5, or CD40, is broadly expressed amongst many cell types, including immune, vascular, and epithelial cells [31]. Ligation of CD40 on APCs supports maturation, which facilitates T-cell activation and differentiation [32]. Inducible T-cell co-stimulator (ICOS) is expressed predominately on activated CD4+ cells. Upon binding to its ligand (ICOSL), ICOS stimulates T-cell proliferation and cytokine production [33]. Recent findings have identified ICOS-mediated phosphoinositide 3-kinase (PI3K) signaling as necessary for transcription factor T-box expressed in T cells (T-bet) expression during anti-CTLA-4 therapy [34]. T-bet regulates commitment to the Th1 lineage and controls IFN-γ production in CD4+ T cells and natural killer cells [35,36]. These data suggest ICOS may be a promising target to heighten Th1-mediated anti-tumor responses. TNFRSF7, or CD27, is highly expressed on the surface of activated T cells and shed into circulation as soluble CD27 [37]. CD27 stimulates the survival of activated T cells throughout the many rounds of division [38]. Agonists of these immune stimulatory pathways have demonstrated synergistic anti-tumor responses when used in conjunction with ICIs prompting clinical trials.

3. Mechanisms of Resistance to Immune Checkpoint Blockade

While anti-CTLA-4 and anti-PD-1/PD-L1 immune checkpoint blockade has shown varying success in cancers, including melanoma, non-small cell lung carcinoma (NSCLC), and urothelial carcinoma, many show low or no response, including gastrointestinal, breast, pancreatic, prostate, sarcoma, and colorectal cancers [39,40,41,42,43,44,45,46,47]. Resistance to immunotherapy is classified as primary resistance in which the cancer is completely refractory or acquired resistance, in which there is initial response yet the cancer relapses and progresses [48]. In this section of the review, we will cover tumor cell-intrinsic and -extrinsic factors that contribute to these forms of immune checkpoint blockade resistance.

3.1. Tumor Mutational Burden

Neoantigens are tumor-specific peptides that result from somatic mutations in cancer cells. It is hypothesized that the number of non-synonymous single nucleotide variants that occur in a tumor is predictive of responsiveness to immune checkpoint blockade therapy. A higher occurrence of these mutations, also referred to as the tumor mutational burden (TMB), increases the likelihood of producing immunogenic peptides that can be recognized by the host immune system. In support of this, deficiencies in DNA damage repair pathways underwrites a hyper-mutational phenotype in which cells accumulate many DNA mutations, and are enriched in tumors of patients with durable responses to anti-PD-1 therapy [40]. Notably, tumors that present microsatellite instability as a result from impaired DNA mismatch repair are especially susceptible to immune checkpoint blockade [49]. Gubin et al. showed that tumor antigen-specific T cells are present in tumors and reactivated with the application of immune checkpoint blockade. Moreover, genomics and bioinformatics were utilized to identify tumor-specific mutant proteins as a primary class of antigens that leads to T cell mediated rejection of tumors [50]. In melanoma, urothelial carcinoma, small cell lung cancer, and NSCLC, high TMB has been shown to be significantly associated with ICI response [40,41,51,52,53,54]. The prognostic value of TMB for immune checkpoint blockade relies on the assumption that there is a direct correlation between the number of neoantigens and robustness of immune infiltration. However, TMB is not predictive of immune infiltration in cancers driven by copy number alterations, such as breast, pancreas, and bladder [55]. Furthermore, even in cancers with a strong association, neoantigen load alone cannot predict ICI response outcome of individual patients. In these cancers, additional stratification based on neoantigen heterogeneity may improve survival predictions. Neoantigens can be presented on all tumor cells (clonal) or in a subpopulation of tumor cells (subclonal). Tumors with high burden of clonal neoantigens were demonstrated to respond to ICIs while poor responders were enriched for subclonal neoantigens [56].

3.2. Tumor Microenvironment

The tumor microenvironment (TME) is a complex milieu of cancer, stromal, and immune cells. Various T-cell subsets have been implicated in modulating response to immunotherapy [57]. Multiple studies have found that the exclusion of CD8+ T cells from the TME is correlated with poor clinical outcome [58,59,60]. The degree of immune infiltration in the TME is generally characterized into three classes: immune-desert, which lacks immune cell infiltration, immune-excluded, which has immune cells in the surrounding stroma (but not penetrating the tumor), and immune-inflamed, which has immune cells adjacent to tumor cells [61,62]. Immune-inflamed or ‘hot’ tumors generally contain immune and tumor cells expressing immune checkpoint molecules and are associated with a more favorable response to ICI therapy [63]. Additionally, a T-cell-inflamed gene signature related to IFN-γ signaling is predictive of anti-PD-1 response across multiple cancers [64]. In opposition, the recruitment of Treg cells, a subset of CD4+ T cells, inhibits anti-tumor immune responses, and is correlated with poor prognosis [65]. Tumor- and stroma-specific signaling pathways have been identified that contribute to T-cell exclusion and therefore resistance to ICI. Tumor cell-specific activation of the WNT β-catenin pathway has been correlated to the absence of T cells in metastatic melanoma and shown to result in T-cell exclusion and ICI resistance in a preclinical model [66]. It is hypothesized that in immune-excluded tumors, stroma contributes to T-cell exclusion by forming a physical extracellular matrix barrier around tumor cells [67]. This is supported by a study that showed inhibition of transforming growth factor-β (TGF-β) in fibroblasts aided T-cell penetration and improved responses to anti-PD-LI therapy in a preclinical model [68]. Furthermore, the extracellular matrix secreted by stroma can influence the migration and positioning of T cells [69]. Cancer-associated fibroblasts (CAFs), the most abundant stromal cell type in tumors, can mediate T-cell death and dysfunction through PD-L2 and FASL [70]. Factors in the TME can thus affect efficacy of ICIs [71].

3.3. Genomic Drivers

Immunoediting is a term that describes the sculpting of tumor cell immunogenicity as the tumor evolves [72]. In ICI-resistant tumors, immunoediting can lead to loss of immunogenic antigens through clonal elimination or chromosomal deletions [73]. This may lead to a tumor that lacks specific neoantigens and can evade immune detection. A crucial step in immune recognition of tumor cells is neoantigen presentation, which occurs through human leukocyte antigen (HLA) class I complexes. Neoantigens may not be presented due to loss of antigen expression or genetic deficiencies in antigen processing or presenting machinery [74,75]. Beta-2-microglobulin (B2M) is important for HLA class I folding and expression at the cell surface. Inactivation of B2M causes defects in antigen presentation, and is enriched in patients that are non-responsive to immune checkpoint blockade [76,77].
A T-cell-inflamed gene signature containing IFN-γ signaling is prognostic of patients that will derive benefit from ICIs [64]. IFN-γ is a cytokine produced by activated T cells and natural killer cells that can inhibit proliferation of targeted cells and enhance immune function through magnification of antigen presentation on APCs. Janus kinase 1 (JAK1) and JAK2 are receptor-associated kinases essential in the initiation of the IFN-γ signaling cascade. Loss-of-function mutations in JAK1 or JAK2 abrogates response to IFN-γ, desensitizing tumor cells to IFN-induced growth inhibition [78]. Furthermore, JAK1/JAK2-deficient tumor cell populations can silence HLA class I antigen presentation, which hinders T-cell recognition [79]. Additionally, cancer cell expression of PD-L1 protects against IFN cytotoxicity through inhibition of signal transducer and activator of transcription 3 (STAT3) phosphorylation, an important transcription factor in the IFN signal transduction pathway [80]. Genomic defects of genes in the IFN-γ pathway have been associated with lack of response to ICI and are implicated in primary and acquired resistance [81,82].
Aberrant signaling through mechanisms, such as mitogen-activated protein kinase (MAPK), PI3K, Wnt, and IFN can negatively affect immune cell recruitment and function; therefore, influence response to ICI therapy [83]. Loss or inactivation of tumor suppressor genes serine/threonine-protein kinase (STK11) and phosphatase and tensin homolog (PTEN) has been associated with poorer response to ICIs and reduced intratumoral T-cell populations [84,85,86]. Intratumoral immune exclusion can also result from activating oncogenic signaling in pathways, such as TGF-β or WNT/β-catenin. TGF-β signaling in the stromal compartment of the TME can induce a myofibroblastic barrier to restrict T cell infiltration, while β-catenin can reduce dendritic cell recruitment and priming of T cells [66,68]. Mutational status of epidermal growth factor receptor (EGFR) has been suggested as a predictor of anti-PD-1 resistance, due to EGFR-mutant positive lung patients demonstrating low response and deriving no overall survival benefit [87,88]. A preclinical model of EGFR-driven lung tumors has shown abnormal signaling can remodel the microenvironment through the expression of immunosuppressive factors to evade immune detection [89].

3.4. Host-Specific Genetic Variation

Response to immune checkpoint blockade relies on T-cell recognition of tumor epitopes to induce anti-tumor activity. The HLA complex is the set of genes encoding cell-surface proteins involved in antigen presentation and are therefore essential in immunosurveillance. Antigens presented on HLA class I are recognized by CD8+ T cells, while antigens presented on HLA class II are recognized by CD4+ T cells. For class I molecules, these peptides are stably bound within a binding pocket consisting of six pockets comprised of polymorphic amino acid residues that establish binding avidity and functionality [90,91,92]. Thousands of functional variants have been observed in the genes, which encode HLA class I molecules, HLA-A, -B, and -C [93]. These variants confer binding of select repertoires of peptides, referred to as the human immunopeptidome. Heterozygosity of HLA-I loci is predicted to result in presentation of a more diverse collection of tumor neoantigens and therefore improved response to ICI [94,95]. Analysis of patients with advanced melanoma or NSCLC treated with immune checkpoint blockade demonstrated homozygosity in at least one HLA-I loci, is associated with reduced survival. Interestingly, this correlation remains significant in the context of mutation load, tumor stage, and drug class [96]. Furthermore, patients with HLA-1 heterozygous loci exhibited increased clonal expansion of TCR repertoires, possibly conferring a stronger T-cell response [96]. In patients, loss of heterozygosity in HLA loci, mutation of HLA genes, and modulation of HLA expression may reduce response to immune checkpoint blockade.
The gut microbiome has been implicated in mediating antitumor immune action and response to ICI. Fecal microbiome samples of patients whom responded to anti-PD-1 treatment presented increased alpha diversity and abundance of Ruminococcaceae bacteria. This profile was associated with stronger antitumor immunity in patients and was recapitulated in a germ-free mouse model [97]. Antibiotic treatment of patients with advanced NSCLC, renal cell carcinoma, or urothelial carcinoma whom received anti-PD-1 therapy is correlated with shorter progression-free survival and overall survival [98]. Quantitative metagenomics of patients before and after anti-PD-1 treatment revealed a higher richness in the composition of the gut microbiota with improved clinical response. In these patients, enrichment of the commensal Akkermansia muciniphila was most associated with responders to immune checkpoint blockade [98].
Disruption of the microbiota can modulate myeloid-derived cell responses in the tumor microenvironment and dampen response to immunotherapy and chemotherapy [99]. These myeloid cells originate from monocytes and granulocytes and are stimulated by tumor-derived factors to remain in activated immature states that may be tumor-promoting. Included in this classification are myeloid-derived suppressor cells (MDSCs), which are defined by their ability to suppress T cells and tumor-associated macrophages (TAMs) [100]. Furthermore, mice fed with Bifidobacterium demonstrated reduced tumor growth and greater intratumoral numbers of CD8+ T cells. Notably, Bifidobacterium administration displayed synergistic anti-tumor responses with anti-PD-L1 therapy [101]. These studies illustrate the influence of the gut microbiota on immune cell function and highlight dysbiosis as in important field in the context of immune checkpoint blockade therapy.

4. Combinations with Immune Checkpoint Inhibitors

Monotherapy ICIs have durable response rates in subsets of patients in many, but not all, cancer types. To extend the efficacy of ICIs to all patients and cancer types, studies exploring synergistic activity with conventional therapies, immune therapies, and small molecule inhibitors are being performed. In addition to offering improved clinical outcomes, these treatments may also offer a more tolerable safety profile for patients with less drug-related adverse events.

4.1. Anti-CTLA-4 and Anti-PD-1

Perhaps unsurprisingly, the combination of anti-CTLA-4 and anti-PD-1 treatments resulted in longer overall survival in patients with advanced melanoma, renal-cell carcinoma, and DNA mismatch repair-deficient/microsatellite instability-high metastatic colorectal cancer [102,103,104]. Though both therapies target immune checkpoints that attenuate T-cell activation, they do so through distinct mechanisms that differentially affect specific T-cell populations [105]. Anti-PD-1 monotherapy results in the expansion of exhausted CD8+ T cells, while dual therapy results in the expansion of activated terminally differentiated effector CD8+ T cells [106]. Anti-CTLA-4 monotherapy increases the expansion of Th1-like CD4+ T cells, while dual therapy further increases the frequency of this population [106,107]. These data confirm that combinational therapies benefit from unique mechanisms of action that cannot be inferred from monotherapies alone. Clinical trials for anti-CTLA-4 and anti-PD-1 combinational therapy have demonstrated promising anti-tumor activity in lung cancers, mesothelioma, esophagogastric cancer, prostate cancer, and sarcoma [108,109,110,111,112,113].

4.2. Chemotherapy, Radiotherapy, and Surgery

Chemotherapy and radiotherapy can sensitize tumor cells to ICIs by increasing immunogenicity following cellular death. The release of tumor antigens and danger-associated molecular patterns (DAMPs) may positively affect immune cell recognition of aberrant cells and prime an efficient immune response [114,115]. This process is referred to as immunogenic cell death (ICD) and is characterized by the translocation of calreticulin (CRT) to the cell surface and release of adenosine triphosphate (ATP) and high mobility group box 1 (HMGB1). Anthracyclines, oxaliplatin, and mafosfamide are able to induce ICD through the production of reactive oxygen species (ROS) and endoplasmic reticulum (ER) stress [116]. Conversely, chemotherapeutics such as cisplatin and mitomycin C are weak inducers of ER stress and do not trigger translocation of CRT and subsequent ICD [117,118]. Additionally, immunosuppressive cells, such as Tregs and MDSCs, are diminished from the TME following treatment, facilitating the infiltration of cytotoxic T cells [119,120,121]. In patients with metastatic NSCLC, improved progression-free survival and overall survival has been observed with the addition of immune checkpoint blockade therapy to chemotherapy [122]. A preclinical model of mesothelioma demonstrated that concomitant treatment with anti-CTLA-4 and gemcitabine resulted in synergistic anti-tumor effect, while phased administration resulted in no significant difference as compared to gemcitabine alone [123]. Clinical data from triple negative breast cancer patients support a short-term induction period of doxorubicin or cisplatin increases the likelihood of response to anti-PD-1, and enriches immune-related genes, including T-cell cytotoxicity and JAK-STAT pathways [124]. Similarly, a phase 2 international study found anti-CTLA-4 treatment following paclitaxel and carboplatin improved immune-related progression-free survival, while concurrent treatment showed no improvement [125]. These studies indicate the timing of immune checkpoint inhibitor administration is key to clinical benefit, likely due to chemotherapy-induced antigen release. Furthermore, concurrent versus phased administration may vary dependent on chemotherapeutic properties and cancers, warranting additional study.
Similar to certain chemotherapeutic agents, radiotherapy has been shown to induce ICD, resulting in phagocytosis of tumor cells, processing of tumor antigens, and priming of CD8+ T cells [126]. In contrast to the pro-immunogenic effects of radiotherapy, there are also immunosuppressive effects from the killing of CD8+ T cells, preservation of Tregs, and activation of TGF-β [127,128]. It is proposed that the combination of ICIs can ameliorate these mechanisms of immunosuppression. In a preclinical model of breast cancer, ionizing radiation and anti-CTLA-4 treatment conferred survival advantage over either alone through the inhibition of lung metastases [129]. Subsequent studies have demonstrated combination with immune checkpoint blockade therapy increases survival in models of glioblastoma multiforme, hepatocellular carcinoma, melanoma, NSCLC, colorectal cancer, and other cancers [130,131,132]. Combined with immune checkpoint inhibitors, radiotherapy can induce antigen-specific immune responses. This was shown to result from increased presence of antigen-experienced T cells and memory T cells. Furthermore, radiotherapy improved antigen cross presentation in lymph nodes, increased intratumoral T cells, and enhanced presentation of tumor-specific antigens [133]. Tumor cells can also adapt to radiotherapy by upregulating expression of PD-L1. In models of melanoma, colorectal, and triple-negative breast cancer, this upregulation resulted from IFN-γ produced by CD8+ T cells. The concurrent administration of anti-PD-L1 treatment was able to increase efficacy of radiotherapy and prevent recurring tumors in long-term surviving cohorts [134]. Additionally, anti-PD-L1 reduces tumor-infiltrating MDSCs through TNF [135]. A phase 3 clinical trial administering anti-PD-L1 following a combination of chemotherapy and radiation therapy showed significantly longer progression-free survival in NSCLC patients [136]. Interestingly, radiotherapy has been observed to have a systemic effect on the reduction of tumors outside the initial treatment field, termed an abscopal effect [137]. These reports have become more frequent with the addition ICIs, with preclinical and clinical data indicating immune checkpoint blockade potentiates abscopal effects [138,139,140,141]. These studies indicate sequencing and timing of radiotherapy and ICIs are likely to affect efficacy. Arguments have been proposed for the administration of immunotherapy prior, concurrent, and subsequent to radiotherapy. Radiotherapy results in the destruction of tumor infiltrating lymphocytes (TILs), which have migrated within tumor margins and demonstrate increased immunological activity towards cancer cells [142]. Therefore, immunotherapy delivered prior to radiotherapy is exerted on increased numbers of TILs and readies the adaptive immune system to respond to antigens released by radiation-induced tumor cell death [143]. Concurrent treatment of immunotherapy and radiotherapy has demonstrated regression of brain metastases in advanced melanoma [144]. When delivered prior to immunotherapy, radiotherapy may maximize immunologic priming—facilitating a stronger immune response to a wider range of epitopes [136,145]. Combinational therapy has demonstrated synergistic anti-tumor effect; however, further study is required to determine optimal timing of treatment.
Major surgery is a common treatment for many solid cancers. While it is nearly impossible to prove the consequence of this, there is some evidence that surgery impacts the rate of systemic tumor cell dissemination into blood vasculature, release of growth factors, and immune suppression [146,147,148,149]. Thus, surgical resection may create a high-risk window during which cells can spread and the host is immunosuppressed. To combat these negative events, ICIs are being explored as neoadjuvant therapies prior to surgery [150]. In preclinical models, blockade of the PD-1/PD-L1 axis post-surgery reduces tumor recurrence and restores dysfunction of CD8+ T cells [151,152]. Moreover, blockade before surgery in the clinical setting has contributed to the expansion of neoantigen-specific T-cell clones in NSCLC patients [153]. Similarly, the administration of CTLA-4 blockade therapy has demonstrated increased activation of T-cell populations. In patients with urothelial carcinoma, treatment with anti-CTLA-4 preoperatively resulted in increased frequency of CD4 + ICOShi T cells, which was then retrospectively shown to correlate with overall survival for metastatic melanoma patients [154]. Furthermore, patients with advanced melanoma exhibited decreased circulating MDSCs and increased intratumoral CD8+ T cells following neoadjuvant anti-CTLA-4 treatment [155].

4.3. Vaccines and Adoptive Cell Therapies

Cancer vaccines utilize tumor-specific antigens to stimulate a specific antitumor immune response but have had few clinical successes. With the development of ICIs and the approval of Sipuleucel-T for the treatment of prostate cancer in 2010, vaccination has become an appealing avenue to increase TILs in poorly immunogenic tumors [156,157]. In preclinical models of poorly immunogenic melanoma and prostate cancer, the combination of anti-CTLA-4 and granulocyte/macrophage colony-stimulating factor (GM-CSF)-expressing vaccine had significant synergistic effects in eradicating tumors and eliciting an immune response dependent on increased CD8+ T cells [158,159]. Interestingly, data indicate optimal benefit is in part dependent on sequence of therapeutic administration, with cohorts receiving anti-CTLA-4 post vaccination exhibiting higher levels of CD8+ T cells and increased tumor-specific lytic function [160]. This additive effect has also been observed in pancreatic and glioma models of cancer, where it was shown that GM-CSF vaccination induced expression of PD-1/PD-L1 and facilitated the action of anti-PD-1 therapy [161,162]. In these models, combinational therapy exerted a significant survival advantage, while neither therapy alone induced benefit. In the clinical setting, anti-CTLA-4 treatment in conjunction with GM-CSF vaccination has demonstrated promising results in pancreatic adenocarcinoma. Dual treated patients demonstrated improved overall survival and enhanced T-cell response [163]. Peptide- and viral-based vaccines have also validated clinical benefit of combinational therapy in human papilloma virus (HPV)-driven cancer and advanced solid cancers [164,165].
Adoptive cell therapy (ACT) is a form of immunotherapy that has achieved durable responses in cancer patients. ACT involves the autologous infusion of TILs or modified immune cells expressing unique T-cell receptors (TCR) or chimeric antigen receptors (CAR), which recognize tumor epitopes to mediate anti-tumor responses [166]. Current protocol for TIL therapy in melanoma patients consists of isolating lymphocytes from resected tumor fragments, ex vivo expansion of populations with tumor cell-killing capacity, and a lymphodepleting preparative regimen prior to reinfusion [167]. While initially most successful in hematological malignancies, ACT has now been established as a powerful treatment option for advanced melanoma and is further being explored in other solid tumors, such as breast and NSCLC [167,168,169,170]. However, challenges to the development of efficacious therapies include lack of tumor-specific antigens, difficulty in penetrating desmoplastic stroma, and an immunosuppressive microenvironment [171,172]. Furthermore, modified lymphocytes can still become exhausted and express immune checkpoints, which hinder activity [173]. The combination of immune checkpoint blockade to ACT in a preclinical model of melanoma has shown that ICIs can aid in the reversion of immunosuppressive TMEs and confer long-term protection against recurrence of solid tumors [174]. Additionally, ICIs can reinvigorate the immune system and support the persistence of CAR T cells in preclinical and clinical models [175,176,177]. Adjuvant immune checkpoint blockade appears to enhance the potency of CAR T-cell therapy and may extend the success observed in hematologic cancers to solid cancers.

4.4. Tumor Microenvironment Factors

Heterogeneity of the TME may largely influence the efficacy of immune checkpoint blockade. Of note are the various populations of immunosuppressive cells, such as MDSCs, tumor-associated macrophages (TAMs), and CAFs that can modulate the immune microenvironment, enhance tumor cell growth, and support immunotherapy resistance. Increased intratumoral presence of immunosuppressive myeloid cells is correlated with poor prognosis and response to immune checkpoint blockade [178,179]. Furthermore, it has been demonstrated that TAMs can capture anti-PD-1 monoclonal antibodies via Fcγ receptors and thereby prevent binding to CD8+ T cells [180]. TAMs are educated by the TME and are broadly classified as M1 or M2 macrophages. M1 macrophages are classically activated macrophages, which produce proinflammatory cytokines and are generally considered anti-tumoral. M2 macrophages produce anti-inflammatory cytokines and inhibit CD8+ T cells. This phenotype is considered pro-tumoral [181]. While T cells are generally the focus when discussing immune checkpoint inhibitors, TAMs also express PD-1. In a preclinical model, blockade of the PD-1/PD-L1 axis enhances macrophage phagocytosis and reduces tumor growth. Furthermore, PD-1+ TAMs expressed a surface profile similar to M2 macrophages [182]. Anti-PD-1 treatment in a model of osteosarcoma pulmonary metastasis resulted in increased migration of M1 macrophages and decreased migration of M2 macrophages [183]. Therapies targeting the interactions and functionality of myeloid cells in the TME have shown promising results in refractory models of cancer. Entinostat is a class-I-selective histone deacetylase (HDAC) inhibitor that improves immune checkpoint therapy by reducing the immunosuppressive ability of MDSCs [184]. Colony-stimulating factor 1 receptor (CSF-1R) blockade results in reduced immune suppression by diminishing the number of TAMs, reprogramming residual TAMs to encourage antigen presentation, and supporting T-cell activation [185]. Focal-adhesion kinase (FAK) is a non-receptor tyrosine kinase, which mediates proliferation, survival, and migration of multiple cell types in the TME. In addition to protecting tumor cells from anoikis, FAK is important for the migration and proliferation of CAFs, and the immunosuppressive capabilities of MDSCs, TAMs, and Tregs [186,187,188]. Inhibition of FAK reduces intratumoral stroma and myeloid cells and potentiates immune checkpoint blockade therapy [189].
Inflammatory mediators secreted by cells in the TME can promote cancer development [190]. Cyclooxygenase (COX) activity has been identified as a driver of immune suppression and inhibitors have demonstrated synergistic effect with immune checkpoint blockade through the partial restoration of tumor immunosurveillance [191,192].

4.5. High-Throughput Methods of Discovering Combinational Therapies

The increasing availability of high-throughput screening technologies has permitted the rational discovery of ICI combinational treatments. Small-molecule and genome-wide libraries have been utilized to identify and target mechanisms, which mediate T-cell interactions with cancer cells. To facilitate time to U.S. FDA approval, multiple studies have highlighted promising combinational therapies through customized screens focused on previously approved medications. Lizotte et al. developed an in vitro luciferase-based screening assay to identify small molecules, which modulate T-cell-mediated killing of tumor cells and validated epidermal growth factor receptor (EGFR) inhibitors at potentiators of anti-PD-1 immunotherapy [193]. Tu et al. utilized in vivo functional genomics to identify discoidin domain receptor 2 (DDR2) as a target mechanism to sensitize multiple tumor types to anti-PD-1 treatment [194]. In more encompassing studies, genome-wide CRISPR-Cas9 screens have been leveraged to further elucidate genes, which permit tumor cell escape from immunosurveillance. Patel et al. utilized an in vitro co-culture method to assemble a comprehensive set of genes whose loss in tumor cells resulted in reduced function of effector CD8+ T cells [195]. Importantly, this study linked their gene set to loss-of-function mutations in immunotherapy refractory patients, thereby proposing novel targets of immune escape for combinational therapies. Recent advances in technology allow the screening of curated CRISPR-Cas9 libraries in innate and adaptive immune populations in vivo to better delineate gene function [196]. Utilizing CHIME, an immune editing system based on chimeric bone marrow, LaFleur et al. were able to implicate the phosphatase PTPN2 in the differentiation of exhausted CD8+ T cells via modulation of type 1 IFN signaling [197].

5. Conclusions

The application of immune checkpoint inhibitors in the treatment of cancer has given us the first real glance at durable responses in patients. This has created a burgeoning field to discover combinations that extend benefits to multiple tumor types and those that are refractory. While the first wave of combinational therapies focused on mechanisms previously implicated in cancer progression, there is now a push to discover novel synergistic treatments via high-throughput screening methods. To derive the most value, there is a necessity to elucidate pathways that allow tumor cells to circumvent immune detection and identify which patients will benefit most from specific combinations.

Funding

The project was supported by CA075115 to D.T.

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Wei, S.C.; Duffy, C.R.; Allison, J.P. Fundamental Mechanisms of Immune Checkpoint Blockade Therapy. Cancer Discov. 2018, 8, 1069–1086. [Google Scholar] [CrossRef] [Green Version]
  2. Dong, C. Helper T Cells and Cancer-Associated Inflammation: A New Direction for Immunotherapy? J. Interferon Cytokine Res. 2017, 37, 383–385. [Google Scholar] [CrossRef] [PubMed]
  3. Annunziato, F.; Cosmi, L.; Liotta, F.; Maggi, E.; Romagnani, S. Human Th1 dichotomy: Origin, phenotype and biologic activities. Immunology 2014. [Google Scholar] [CrossRef] [Green Version]
  4. Darvin, P.; Toor, S.M.; Sasidharan Nair, V.; Elkord, E. Immune checkpoint inhibitors: Recent progress and potential biomarkers. Exp. Mol. Med. 2018, 50, 165. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  5. Rudd, C.E.; Taylor, A.; Schneider, H. CD28 and CTLA-4 coreceptor expression and signal transduction. Immunol. Rev. 2009, 229, 12–26. [Google Scholar] [CrossRef] [Green Version]
  6. Egen, J.G.; Allison, J.P. Cytotoxic T lymphocyte antigen-4 accumulation in the immunological synapse is regulated by TCR signal strength. Immunity 2002, 16, 23–35. [Google Scholar] [CrossRef] [Green Version]
  7. Qureshi, O.S.; Zheng, Y.; Nakamura, K.; Attridge, K.; Manzotti, C.; Schmidt, E.M.; Baker, J.; Jeffery, L.E.; Kaur, S.; Briggs, Z.; et al. Trans-endocytosis of CD80 and CD86: A molecular basis for the cell-extrinsic function of CTLA-4. Science 2011, 332, 600–603. [Google Scholar] [CrossRef] [Green Version]
  8. Ramagopal, U.A.; Liu, W.; Garrett-Thomson, S.C.; Bonanno, J.B.; Yan, Q.; Srinivasan, M.; Wong, S.C.; Bell, A.; Mankikar, S.; Rangan, V.S.; et al. Structural basis for cancer immunotherapy by the first-in-class checkpoint inhibitor ipilimumab. Proc. Natl. Acad. Sci. USA 2017, 114, E4223–E4232. [Google Scholar] [CrossRef] [Green Version]
  9. 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]
  10. Taube, J.M.; Young, G.D.; McMiller, T.L.; Chen, S.; Salas, J.T.; Pritchard, T.S.; Xu, H.; Meeker, A.K.; Fan, J.; Cheadle, C.; et al. Differential Expression of Immune-Regulatory Genes Associated with PD-L1 Display in Melanoma: Implications for PD-1 Pathway Blockade. Clin. Cancer Res. 2015, 21, 3969–3976. [Google Scholar] [CrossRef] [Green Version]
  11. Juneja, V.R.; McGuire, K.A.; Manguso, R.T.; LaFleur, M.W.; Collins, N.; Haining, W.N.; Freeman, G.J.; Sharpe, A.H. PD-L1 on tumor cells is sufficient for immune evasion in immunogenic tumors and inhibits CD8 T cell cytotoxicity. J. Exp. Med. 2017, 214, 895–904. [Google Scholar] [CrossRef]
  12. Latchman, Y.; Wood, C.R.; Chernova, T.; Chaudhary, D.; Borde, M.; Chernova, I.; Iwai, Y.; Long, A.J.; Brown, J.A.; Nunes, R.; et al. PD-L2 is a second ligand for PD-1 and inhibits T cell activation. Nat. Immunol. 2001, 2, 261–268. [Google Scholar] [CrossRef] [PubMed]
  13. Sidorenko, S.P.; Clark, E.A. The dual-function CD150 receptor subfamily: The viral attraction. Nat. Immunol. 2003, 4, 19–24. [Google Scholar] [CrossRef] [PubMed]
  14. Hui, E.; Cheung, J.; Zhu, J.; Su, X.; Taylor, M.J.; Wallweber, H.A.; Sasmal, D.K.; Huang, J.; Kim, J.M.; Mellman, I.; et al. T cell costimulatory receptor CD28 is a primary target for PD-1-mediated inhibition. Science 2017, 355, 1428–1433. [Google Scholar] [CrossRef] [PubMed]
  15. Sheppard, K.A.; Fitz, L.J.; Lee, J.M.; Benander, C.; George, J.A.; Wooters, J.; Qiu, Y.; Jussif, J.M.; Carter, L.L.; Wood, C.R.; et al. PD-1 inhibits T-cell receptor induced phosphorylation of the ZAP70/CD3zeta signalosome and downstream signaling to PKCtheta. FEBS Lett. 2004, 574, 37–41. [Google Scholar] [CrossRef] [Green Version]
  16. Pauken, K.E.; Wherry, E.J. Overcoming T cell exhaustion in infection and cancer. Trends Immunol. 2015, 36, 265–276. [Google Scholar] [CrossRef] [Green Version]
  17. Huard, B.; Prigent, P.; Tournier, M.; Bruniquel, D.; Triebel, F. CD4/major histocompatibility complex class II interaction analyzed with CD4- and lymphocyte activation gene-3 (LAG-3)-Ig fusion proteins. Eur. J. Immunol. 1995, 25, 2718–2721. [Google Scholar] [CrossRef]
  18. Workman, C.J.; Cauley, L.S.; Kim, I.J.; Blackman, M.A.; Woodland, D.L.; Vignali, D.A. Lymphocyte activation gene-3 (CD223) regulates the size of the expanding T cell population following antigen activation in vivo. J. Immunol. 2004, 172, 5450–5455. [Google Scholar] [CrossRef]
  19. Zhu, C.; Anderson, A.C.; Schubart, A.; Xiong, H.; Imitola, J.; Khoury, S.J.; Zheng, X.X.; Strom, T.B.; Kuchroo, V.K. The Tim-3 ligand galectin-9 negatively regulates T helper type 1 immunity. Nat. Immunol. 2005, 6, 1245–1252. [Google Scholar] [CrossRef]
  20. Stanietsky, N.; Rovis, T.L.; Glasner, A.; Seidel, E.; Tsukerman, P.; Yamin, R.; Enk, J.; Jonjic, S.; Mandelboim, O. Mouse TIGIT inhibits NK-cell cytotoxicity upon interaction with PVR. Eur. J. Immunol. 2013, 43, 2138–2150. [Google Scholar] [CrossRef]
  21. Yu, X.; Harden, K.; Gonzalez, L.C.; Francesco, M.; Chiang, E.; Irving, B.; Tom, I.; Ivelja, S.; Refino, C.J.; Clark, H.; et al. The surface protein TIGIT suppresses T cell activation by promoting the generation of mature immunoregulatory dendritic cells. Nat. Immunol. 2009, 10, 48–57. [Google Scholar] [CrossRef]
  22. Rogers, P.R.; Song, J.; Gramaglia, I.; Killeen, N.; Croft, M. OX40 promotes Bcl-xL and Bcl-2 expression and is essential for long-term survival of CD4 T cells. Immunity 2001, 15, 445–455. [Google Scholar] [CrossRef] [Green Version]
  23. Shimizu, J.; Yamazaki, S.; Takahashi, T.; Ishida, Y.; Sakaguchi, S. Stimulation of CD25(+)CD4(+) regulatory T cells through GITR breaks immunological self-tolerance. Nat. Immunol. 2002, 3, 135–142. [Google Scholar] [CrossRef]
  24. Schaer, D.A.; Murphy, J.T.; Wolchok, J.D. Modulation of GITR for cancer immunotherapy. Curr. Opin. Immunol. 2012, 24, 217–224. [Google Scholar] [CrossRef] [Green Version]
  25. Gurney, A.L.; Marsters, S.A.; Huang, R.M.; Pitti, R.M.; Mark, D.T.; Baldwin, D.T.; Gray, A.M.; Dowd, A.D.; Brush, A.D.; Heldens, A.D.; et al. Identification of a new member of the tumor necrosis factor family and its receptor, a human ortholog of mouse GITR. Curr. Biol. 1999, 9, 215–218. [Google Scholar] [CrossRef] [Green Version]
  26. Côté, A.L.; Zhang, P.; O’Sullivan, J.A.; Jacobs, V.L.; Clemis, C.R.; Sakaguchi, S.; Guevara-Patiño, J.A.; Turk, M.J. Stimulation of the glucocorticoid-induced TNF receptor family-related receptor on CD8 T cells induces protective and high-avidity T cell responses to tumor-specific antigens. J. Immunol. 2011, 186, 275–283. [Google Scholar] [CrossRef] [Green Version]
  27. Ronchetti, S.; Zollo, O.; Bruscoli, S.; Agostini, M.; Bianchini, R.; Nocentini, G.; Ayroldi, E.; Riccardi, C. GITR, a member of the TNF receptor superfamily, is costimulatory to mouse T lymphocyte subpopulations. Eur. J. Immunol. 2004, 34, 613–622. [Google Scholar] [CrossRef]
  28. McHugh, R.S.; Whitters, M.J.; Piccirillo, C.A.; Young, D.A.; Shevach, E.M.; Collins, M.; Byrne, M.C. CD4(+)CD25(+) immunoregulatory T cells: Gene expression analysis reveals a functional role for the glucocorticoid-induced TNF receptor. Immunity 2002, 16, 311–323. [Google Scholar] [CrossRef] [Green Version]
  29. Shuford, W.W.; Klussman, K.; Tritchler, D.D.; Loo, D.T.; Chalupny, J.; Siadak, A.W.; Brown, T.J.; Emswiler, J.; Raecho, H.; Larsen, C.P.; et al. 4-1BB costimulatory signals preferentially induce CD8+ T cell proliferation and lead to the amplification in vivo of cytotoxic T cell responses. J. Exp. Med. 1997, 186, 47–55. [Google Scholar] [CrossRef]
  30. Hernandez-Chacon, J.A.; Li, Y.; Wu, R.C.; Bernatchez, C.; Wang, Y.; Weber, J.S.; Hwu, P.; Radvanyi, L.G. Costimulation through the CD137/4-1BB pathway protects human melanoma tumor-infiltrating lymphocytes from activation-induced cell death and enhances antitumor effector function. J. Immunother. 2011, 34, 236–250. [Google Scholar] [CrossRef] [Green Version]
  31. Vonderheide, R.H. Prospect of targeting the CD40 pathway for cancer therapy. Clin. Cancer Res. 2007, 13, 1083–1088. [Google Scholar] [CrossRef] [Green Version]
  32. Elgueta, R.; Benson, M.J.; de Vries, V.C.; Wasiuk, A.; Guo, Y.; Noelle, R.J. Molecular mechanism and function of CD40/CD40L engagement in the immune system. Immunol. Rev. 2009, 229, 152–172. [Google Scholar] [CrossRef] [Green Version]
  33. Hutloff, A.; Dittrich, A.M.; Beier, K.C.; Eljaschewitsch, B.; Kraft, R.; Anagnostopoulos, I.; Kroczek, R.A. ICOS is an inducible T-cell co-stimulator structurally and functionally related to CD28. Nature 1999, 397, 263–266. [Google Scholar] [CrossRef]
  34. Chen, H.; Fu, T.; Suh, W.K.; Tsavachidou, D.; Wen, S.; Gao, J.; Ng Tang, D.; He, Q.; Sun, J.; Sharma, P. CD4 T cells require ICOS-mediated PI3K signaling to increase T-Bet expression in the setting of anti-CTLA-4 therapy. Cancer Immunol. Res. 2014, 2, 167–176. [Google Scholar] [CrossRef] [Green Version]
  35. Szabo, S.J.; Kim, S.T.; Costa, G.L.; Zhang, X.; Fathman, C.G.; Glimcher, L.H. A novel transcription factor, T-bet, directs Th1 lineage commitment. Cell 2000, 100, 655–669. [Google Scholar] [CrossRef] [Green Version]
  36. Szabo, S.J.; Sullivan, B.M.; Stemmann, C.; Satoskar, A.R.; Sleckman, B.P.; Glimcher, L.H. Distinct effects of T-bet in TH1 lineage commitment and IFN-gamma production in CD4 and CD8 T cells. Science 2002, 295, 338–342. [Google Scholar] [CrossRef]
  37. Lens, S.M.; Tesselaar, K.; van Oers, M.H.; van Lier, R.A. Control of lymphocyte function through CD27-CD70 interactions. Semin. Immunol. 1998, 10, 491–499. [Google Scholar] [CrossRef]
  38. Hendriks, J.; Xiao, Y.; Borst, J. CD27 promotes survival of activated T cells and complements CD28 in generation and establishment of the effector T cell pool. J. Exp. Med. 2003, 198, 1369–1380. [Google Scholar] [CrossRef] [Green Version]
  39. 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]
  40. 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] [Green Version]
  41. Rosenberg, J.E.; Hoffman-Censits, J.; Powles, T.; van der Heijden, M.S.; Balar, A.V.; Necchi, A.; Dawson, N.; O’Donnell, P.H.; Balmanoukian, A.; Loriot, Y.; et al. Atezolizumab in patients with locally advanced and metastatic urothelial carcinoma who have progressed following treatment with platinum-based chemotherapy: A single-arm, multicentre, phase 2 trial. Lancet 2016, 387, 1909–1920. [Google Scholar] [CrossRef] [Green Version]
  42. Shitara, K.; Özgüroğlu, M.; Bang, Y.J.; Di Bartolomeo, M.; Mandalà, M.; Ryu, M.H.; Fornaro, L.; Olesiński, T.; Caglevic, C.; Chung, H.C.; et al. Pembrolizumab versus paclitaxel for previously treated, advanced gastric or gastro-oesophageal junction cancer (KEYNOTE-061): A randomised, open-label, controlled, phase 3 trial. Lancet 2018, 392, 123–133. [Google Scholar] [CrossRef]
  43. Adams, S.; Schmid, P.; Rugo, H.S.; Winer, E.P.; Loirat, D.; Awada, A.; Cescon, D.W.; Iwata, H.; Campone, M.; Nanda, R.; et al. Pembrolizumab monotherapy for previously treated metastatic triple-negative breast cancer: Cohort A of the phase II KEYNOTE-086 study. Ann. Oncol. 2019, 30, 397–404. [Google Scholar] [CrossRef] [Green Version]
  44. O’Reilly, E.M.; Oh, D.Y.; Dhani, N.; Renouf, D.J.; Lee, M.A.; Sun, W.; Fisher, G.; Hezel, A.; Chang, S.C.; Vlahovic, G.; et al. Durvalumab With or Without Tremelimumab for Patients With Metastatic Pancreatic Ductal Adenocarcinoma: A Phase 2 Randomized Clinical Trial. JAMA Oncol. 2019. [Google Scholar] [CrossRef]
  45. Antonarakis, E.S.; Piulats, J.M.; Gross-Goupil, M.; Goh, J.; Ojamaa, K.; Hoimes, C.J.; Vaishampayan, U.; Berger, R.; Sezer, A.; Alanko, T.; et al. Pembrolizumab for Treatment-Refractory Metastatic Castration-Resistant Prostate Cancer: Multicohort, Open-Label Phase II KEYNOTE-199 Study. J. Clin. Oncol. 2019, 38, 395–405. [Google Scholar] [CrossRef]
  46. Maki, R.G.; Jungbluth, A.A.; Gnjatic, S.; Schwartz, G.K.; D’Adamo, D.R.; Keohan, M.L.; Wagner, M.J.; Scheu, K.; Chiu, R.; Ritter, E.; et al. A Pilot Study of Anti-CTLA4 Antibody Ipilimumab in Patients with Synovial Sarcoma. Sarcoma 2013, 2013, 168145. [Google Scholar] [CrossRef]
  47. Le, D.T.; Uram, J.N.; Wang, H.; Bartlett, B.R.; Kemberling, H.; Eyring, A.D.; Skora, A.D.; Luber, B.S.; Azad, N.S.; Laheru, D.; et al. PD-1 Blockade in Tumors with Mismatch-Repair Deficiency. N. Engl. J. Med. 2015, 372, 2509–2520. [Google Scholar] [CrossRef]
  48. Sharma, P.; Hu-Lieskovan, S.; Wargo, J.A.; Ribas, A. Primary, Adaptive, and Acquired Resistance to Cancer Immunotherapy. Cell 2017, 168, 707–723. [Google Scholar] [CrossRef] [Green Version]
  49. Le, D.T.; Durham, J.N.; Smith, K.N.; Wang, H.; Bartlett, B.R.; Aulakh, L.K.; Lu, S.; Kemberling, H.; Wilt, C.; Luber, B.S.; et al. Mismatch repair deficiency predicts response of solid tumors to PD-1 blockade. Science 2017, 357, 409–413. [Google Scholar] [CrossRef] [Green Version]
  50. Gubin, M.M.; Zhang, X.; Schuster, H.; Caron, E.; Ward, J.P.; Noguchi, T.; Ivanova, Y.; Hundal, J.; Arthur, C.D.; Krebber, W.J.; et al. Checkpoint blockade cancer immunotherapy targets tumour-specific mutant antigens. Nature 2014, 515, 577–581. [Google Scholar] [CrossRef]
  51. Snyder, A.; Makarov, V.; Merghoub, T.; Yuan, J.; Zaretsky, J.M.; Desrichard, A.; Walsh, L.A.; Postow, M.A.; Wong, P.; Ho, T.S.; et al. Genetic basis for clinical response to CTLA-4 blockade in melanoma. N. Engl. J. Med. 2014, 371, 2189–2199. [Google Scholar] [CrossRef]
  52. Van Allen, E.M.; Miao, D.; Schilling, B.; Shukla, S.A.; Blank, C.; Zimmer, L.; Sucker, A.; Hillen, U.; Foppen, M.H.G.; Goldinger, S.M.; et al. Genomic correlates of response to CTLA-4 blockade in metastatic melanoma. Science 2015, 350, 207–211. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  53. Hellmann, M.D.; Callahan, M.K.; Awad, M.M.; Calvo, E.; Ascierto, P.A.; Atmaca, A.; Rizvi, N.A.; Hirsch, F.R.; Selvaggi, G.; Szustakowski, J.D.; et al. Tumor Mutational Burden and Efficacy of Nivolumab Monotherapy and in Combination with Ipilimumab in Small-Cell Lung Cancer. Cancer Cell 2018, 33, 853–861. [Google Scholar] [CrossRef] [PubMed]
  54. Hellmann, M.D.; Nathanson, T.; Rizvi, H.; Creelan, B.C.; Sanchez-Vega, F.; Ahuja, A.; Ni, A.; Novik, J.B.; Mangarin, L.M.B.; Abu-Akeel, M.; et al. Genomic Features of Response to Combination Immunotherapy in Patients with Advanced Non-Small-Cell Lung Cancer. Cancer Cell 2018, 33, 843–852. [Google Scholar] [CrossRef] [Green Version]
  55. McGrail, D.J.; Federico, L.; Li, Y.; Dai, H.; Lu, Y.; Mills, G.B.; Yi, S.; Lin, S.Y.; Sahni, N. Multi-omics analysis reveals neoantigen-independent immune cell infiltration in copy-number driven cancers. Nat. Commun. 2018, 9, 1317. [Google Scholar] [CrossRef] [Green Version]
  56. McGranahan, N.; Furness, A.J.; Rosenthal, R.; Ramskov, S.; Lyngaa, R.; Saini, S.K.; Jamal-Hanjani, M.; Wilson, G.A.; Birkbak, N.J.; Hiley, C.T.; et al. Clonal neoantigens elicit T cell immunoreactivity and sensitivity to immune checkpoint blockade. Science 2016, 351, 1463–1469. [Google Scholar] [CrossRef] [Green Version]
  57. Chraa, D.; Naim, A.; Olive, D.; Badou, A. T lymphocyte subsets in cancer immunity: Friends or foes. J. Leukoc. Biol. 2019, 105, 243–255. [Google Scholar] [CrossRef]
  58. Galon, J.; Costes, A.; Sanchez-Cabo, F.; Kirilovsky, A.; Mlecnik, B.; Lagorce-Pagès, C.; Tosolini, M.; Camus, M.; Berger, A.; Wind, P.; et al. Type, density, and location of immune cells within human colorectal tumors predict clinical outcome. Science 2006, 313, 1960–1964. [Google Scholar] [CrossRef] [Green Version]
  59. Pagès, F.; Berger, A.; Camus, M.; Sanchez-Cabo, F.; Costes, A.; Molidor, R.; Mlecnik, B.; Kirilovsky, A.; Nilsson, M.; Damotte, D.; et al. Effector memory T cells, early metastasis, and survival in colorectal cancer. N. Engl. J. Med. 2005, 353, 2654–2666. [Google Scholar] [CrossRef]
  60. Sato, E.; Olson, S.H.; Ahn, J.; Bundy, B.; Nishikawa, H.; Qian, F.; Jungbluth, A.A.; Frosina, D.; Gnjatic, S.; Ambrosone, C.; et al. Intraepithelial CD8+ tumor-infiltrating lymphocytes and a high CD8+/regulatory T cell ratio are associated with favorable prognosis in ovarian cancer. Proc. Natl. Acad. Sci. USA 2005, 102, 18538–18543. [Google Scholar] [CrossRef] [Green Version]
  61. Chen, D.S.; Mellman, I. Elements of cancer immunity and the cancer-immune set point. Nature 2017, 541, 321–330. [Google Scholar] [CrossRef]
  62. Lanitis, E.; Dangaj, D.; Irving, M.; Coukos, G. Mechanisms regulating T-cell infiltration and activity in solid tumors. Ann. Oncol. 2017, 28, xii18–xii32. [Google Scholar] [CrossRef] [PubMed]
  63. 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] [Green Version]
  64. Ayers, M.; Lunceford, J.; Nebozhyn, M.; Murphy, E.; Loboda, A.; Kaufman, D.R.; Albright, A.; Cheng, J.D.; Kang, S.P.; Shankaran, V.; et al. IFN-γ-related mRNA profile predicts clinical response to PD-1 blockade. J. Clin. Investig. 2017, 127, 2930–2940. [Google Scholar] [CrossRef] [PubMed]
  65. Yano, H.; Andrews, L.P.; Workman, C.J.; Vignali, D.A.A. Intratumoral regulatory T cells: Markers, subsets and their impact on anti-tumor immunity. Immunology 2019, 157, 232–247. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  66. Spranger, S.; Bao, R.; Gajewski, T.F. Melanoma-intrinsic β-catenin signalling prevents anti-tumour immunity. Nature 2015, 523, 231–235. [Google Scholar] [CrossRef]
  67. Menon, A.G.; Fleuren, G.J.; Alphenaar, E.A.; Jonges, L.E.; Janssen van Rhijn, C.M.; Ensink, N.G.; Putter, H.; Tollenaar, R.A.; van de Velde, C.J.; Kuppen, P.J. A basal membrane-like structure surrounding tumour nodules may prevent intraepithelial leucocyte infiltration in colorectal cancer. Cancer Immunol. Immunother. 2003, 52, 121–126. [Google Scholar] [CrossRef]
  68. Mariathasan, S.; Turley, S.J.; Nickles, D.; Castiglioni, A.; Yuen, K.; Wang, Y.; Kadel, E.E.; Koeppen, H.; Astarita, J.L.; Cubas, R.; et al. TGFβ attenuates tumour response to PD-L1 blockade by contributing to exclusion of T cells. Nature 2018, 554, 544–548. [Google Scholar] [CrossRef]
  69. Salmon, H.; Franciszkiewicz, K.; Damotte, D.; Dieu-Nosjean, M.C.; Validire, P.; Trautmann, A.; Mami-Chouaib, F.; Donnadieu, E. Matrix architecture defines the preferential localization and migration of T cells into the stroma of human lung tumors. J. Clin. Investig. 2012, 122, 899–910. [Google Scholar] [CrossRef] [Green Version]
  70. Lakins, M.A.; Ghorani, E.; Munir, H.; Martins, C.P.; Shields, J.D. Cancer-associated fibroblasts induce antigen-specific deletion of CD8. Nat. Commun. 2018, 9, 948. [Google Scholar] [CrossRef]
  71. Becht, E.; Giraldo, N.A.; Dieu-Nosjean, M.C.; Sautès-Fridman, C.; Fridman, W.H. Cancer immune contexture and immunotherapy. Curr. Opin. Immunol. 2016, 39, 7–13. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  72. Dunn, G.P.; Bruce, A.T.; Ikeda, H.; Old, L.J.; Schreiber, R.D. Cancer immunoediting: From immunosurveillance to tumor escape. Nat. Immunol. 2002, 3, 991–998. [Google Scholar] [CrossRef] [PubMed]
  73. 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] [Green Version]
  74. Marincola, F.M.; Jaffee, E.M.; Hicklin, D.J.; Ferrone, S. Escape of human solid tumors from T-cell recognition: Molecular mechanisms and functional significance. Adv. Immunol. 2000, 74, 181–273. [Google Scholar] [PubMed]
  75. Sucker, A.; Zhao, F.; Real, B.; Heeke, C.; Bielefeld, N.; Maβen, S.; Horn, S.; Moll, I.; Maltaner, R.; Horn, P.A.; et al. Genetic evolution of T-cell resistance in the course of melanoma progression. Clin. Cancer Res. 2014, 20, 6593–6604. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  76. Sade-Feldman, M.; Jiao, Y.J.; Chen, J.H.; Rooney, M.S.; Barzily-Rokni, M.; Eliane, J.P.; Bjorgaard, S.L.; Hammond, M.R.; Vitzthum, H.; Blackmon, S.M.; et al. Resistance to checkpoint blockade therapy through inactivation of antigen presentation. Nat. Commun. 2017, 8, 1136. [Google Scholar] [CrossRef] [PubMed]
  77. Gettinger, S.; Choi, J.; Hastings, K.; Truini, A.; Datar, I.; Sowell, R.; Wurtz, A.; Dong, W.; Cai, G.; Melnick, M.A.; et al. Impaired HLA Class I Antigen Processing and Presentation as a Mechanism of Acquired Resistance to Immune Checkpoint Inhibitors in Lung Cancer. Cancer Discov. 2017, 7, 1420–1435. [Google Scholar] [CrossRef] [Green Version]
  78. Zaretsky, J.M.; Garcia-Diaz, A.; Shin, D.S.; Escuin-Ordinas, H.; Hugo, W.; Hu-Lieskovan, S.; Torrejon, D.Y.; Abril-Rodriguez, G.; Sandoval, S.; Barthly, L.; et al. Mutations Associated with Acquired Resistance to PD-1 Blockade in Melanoma. N. Engl. J. Med. 2016, 375, 819–829. [Google Scholar] [CrossRef]
  79. Sucker, A.; Zhao, F.; Pieper, N.; Heeke, C.; Maltaner, R.; Stadtler, N.; Real, B.; Bielefeld, N.; Howe, S.; Weide, B.; et al. Acquired IFNγ resistance impairs anti-tumor immunity and gives rise to T-cell-resistant melanoma lesions. Nat. Commun. 2017, 8, 15440. [Google Scholar] [CrossRef]
  80. Gato-Cañas, M.; Zuazo, M.; Arasanz, H.; Ibañez-Vea, M.; Lorenzo, L.; Fernandez-Hinojal, G.; Vera, R.; Smerdou, C.; Martisova, E.; Arozarena, I.; et al. PDL1 Signals through Conserved Sequence Motifs to Overcome Interferon-Mediated Cytotoxicity. Cell Rep. 2017, 20, 1818–1829. [Google Scholar] [CrossRef] [Green Version]
  81. Gao, J.; Shi, L.Z.; Zhao, H.; Chen, J.; Xiong, L.; He, Q.; Chen, T.; Roszik, J.; Bernatchez, C.; Woodman, S.E.; et al. Loss of IFN-γ Pathway Genes in Tumor Cells as a Mechanism of Resistance to Anti-CTLA-4 Therapy. Cell 2016, 167, 397–404. [Google Scholar] [CrossRef] [PubMed]
  82. 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] [PubMed] [Green Version]
  83. Spranger, S.; Gajewski, T.F. Impact of oncogenic pathways on evasion of antitumour immune responses. Nat. Rev. Cancer 2018, 18, 139–147. [Google Scholar] [CrossRef] [PubMed]
  84. Rizvi, H.; Sanchez-Vega, F.; La, K.; Chatila, W.; Jonsson, P.; Halpenny, D.; Plodkowski, A.; Long, N.; Sauter, J.L.; Rekhtman, N.; et al. Molecular Determinants of Response to Anti-Programmed Cell Death (PD)-1 and Anti-Programmed Death-Ligand 1 (PD-L1) Blockade in Patients With Non-Small-Cell Lung Cancer Profiled With Targeted Next-Generation Sequencing. J. Clin. Oncol. 2018, 36, 633–641. [Google Scholar] [CrossRef]
  85. 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]
  86. Peng, W.; Chen, J.Q.; Liu, C.; Malu, S.; Creasy, C.; Tetzlaff, M.T.; Xu, C.; McKenzie, J.A.; Zhang, C.; Liang, X.; et al. Loss of PTEN Promotes Resistance to T Cell-Mediated Immunotherapy. Cancer Discov. 2016, 6, 202–216. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  87. Gainor, J.F.; Shaw, A.T.; Sequist, L.V.; Fu, X.; Azzoli, C.G.; Piotrowska, Z.; Huynh, T.G.; Zhao, L.; Fulton, L.; Schultz, K.R.; et al. EGFR Mutations and ALK Rearrangements Are Associated with Low Response Rates to PD-1 Pathway Blockade in Non-Small Cell Lung Cancer: A Retrospective Analysis. Clin. Cancer Res. 2016, 22, 4585–4593. [Google Scholar] [CrossRef] [Green Version]
  88. Lee, C.K.; Man, J.; Lord, S.; Cooper, W.; Links, M.; Gebski, V.; Herbst, R.S.; Gralla, R.J.; Mok, T.; Yang, J.C. Clinical and Molecular Characteristics Associated With Survival Among Patients Treated With Checkpoint Inhibitors for Advanced Non-Small Cell Lung Carcinoma: A Systematic Review and Meta-analysis. JAMA Oncol. 2018, 4, 210–216. [Google Scholar] [CrossRef]
  89. Akbay, E.A.; Koyama, S.; Carretero, J.; Altabef, A.; Tchaicha, J.H.; Christensen, C.L.; Mikse, O.R.; Cherniack, A.D.; Beauchamp, E.M.; Pugh, T.J.; et al. Activation of the PD-1 pathway contributes to immune escape in EGFR-driven lung tumors. Cancer Discov. 2013, 3, 1355–1363. [Google Scholar] [CrossRef] [Green Version]
  90. Matsumura, M.; Fremont, D.H.; Peterson, P.A.; Wilson, I.A. Emerging principles for the recognition of peptide antigens by MHC class I molecules. Science 1992, 257, 927–934. [Google Scholar] [CrossRef]
  91. Van Deutekom, H.W.; Keşmir, C. Zooming into the binding groove of HLA molecules: Which positions and which substitutions change peptide binding most? Immunogenetics 2015, 67, 425–436. [Google Scholar] [CrossRef] [Green Version]
  92. Zhao, B.; Png, A.E.; Ren, E.C.; Kolatkar, P.R.; Mathura, V.S.; Sakharkar, M.K.; Kangueane, P. Compression of functional space in HLA-A sequence diversity. Hum. Immunol. 2003, 64, 718–728. [Google Scholar] [CrossRef]
  93. Robinson, J.; Halliwell, J.A.; Hayhurst, J.D.; Flicek, P.; Parham, P.; Marsh, S.G. The IPD and IMGT/HLA database: Allele variant databases. Nucleic Acids Res. 2015, 43, D423–D431. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  94. Doherty, P.C.; Zinkernagel, R.M. A biological role for the major histocompatibility antigens. Lancet 1975, 1, 1406–1409. [Google Scholar] [CrossRef]
  95. Aptsiauri, N.; Cabrera, T.; Garcia-Lora, A.; Lopez-Nevot, M.A.; Ruiz-Cabello, F.; Garrido, F. MHC class I antigens and immune surveillance in transformed cells. Int. Rev. Cytol. 2007, 256, 139–189. [Google Scholar] [CrossRef]
  96. Chowell, D.; Morris, L.G.T.; Grigg, C.M.; Weber, J.K.; Samstein, R.M.; Makarov, V.; Kuo, F.; Kendall, S.M.; Requena, D.; Riaz, N.; et al. Patient HLA class I genotype influences cancer response to checkpoint blockade immunotherapy. Science 2018, 359, 582–587. [Google Scholar] [CrossRef] [Green Version]
  97. 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] [PubMed] [Green Version]
  98. Routy, B.; Le Chatelier, E.; Derosa, L.; Duong, C.P.M.; Alou, M.T.; Daillere, 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]
  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. Awad, R.M.; De Vlaeminck, Y.; Maebe, J.; Goyvaerts, C.; Breckpot, K. Turn Back the TIMe: Targeting Tumor Infiltrating Myeloid Cells to Revert Cancer Progression. Front. Immunol. 2018, 9, 1977. [Google Scholar] [CrossRef]
  101. Sivan, A.; Corrales, L.; Hubert, N.; Williams, J.B.; Aquino-Michaels, K.; Earley, Z.M.; Benyamin, F.W.; Lei, Y.M.; Jabri, B.; Alegre, M.L.; et al. Commensal Bifidobacterium promotes antitumor immunity and facilitates anti-PD-L1 efficacy. Science 2015, 350, 1084–1089. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  102. Wolchok, J.D.; Chiarion-Sileni, V.; Gonzalez, R.; Rutkowski, P.; Grob, J.J.; Cowey, C.L.; Lao, C.D.; Wagstaff, J.; Schadendorf, D.; Ferrucci, P.F.; et al. Overall Survival with Combined Nivolumab and Ipilimumab in Advanced Melanoma. N. Engl. J. Med. 2017, 377, 1345–1356. [Google Scholar] [CrossRef] [PubMed]
  103. Motzer, R.J.; Tannir, N.M.; McDermott, D.F.; Arén Frontera, O.; Melichar, B.; Choueiri, T.K.; Plimack, E.R.; Barthélémy, P.; Porta, C.; George, S.; et al. Nivolumab plus Ipilimumab versus Sunitinib in Advanced Renal-Cell Carcinoma. N. Engl. J. Med. 2018, 378, 1277–1290. [Google Scholar] [CrossRef] [PubMed]
  104. Overman, M.J.; Lonardi, S.; Wong, K.Y.M.; Lenz, H.J.; Gelsomino, F.; Aglietta, M.; Morse, M.A.; Van Cutsem, E.; McDermott, R.; Hill, A.; et al. Durable Clinical Benefit With Nivolumab Plus Ipilimumab in DNA Mismatch Repair-Deficient/Microsatellite Instability-High Metastatic Colorectal Cancer. J. Clin. Oncol. 2018, 36, 773–779. [Google Scholar] [CrossRef] [PubMed]
  105. Das, R.; Verma, R.; Sznol, M.; Boddupalli, C.S.; Gettinger, S.N.; Kluger, H.; Callahan, M.; Wolchok, J.D.; Halaban, R.; Dhodapkar, M.V.; et al. Combination therapy with anti-CTLA-4 and anti-PD-1 leads to distinct immunologic changes in vivo. J. Immunol. 2015, 194, 950–959. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  106. Wei, S.C.; Anang, N.-A.A.S.; Sharma, R.; Andrews, M.C.; Reuben, A.; Levine, J.H.; Cogdill, A.P.; Mancuso, J.J.; Wargo, J.A.; Pe’er, D.; et al. Combination anti–CTLA-4 plus anti–PD-1 checkpoint blockade utilizes cellular mechanisms partially distinct from monotherapies. Proc. Natl. Acad. Sci. USA 2019. [Google Scholar] [CrossRef] [Green Version]
  107. Wei, S.C.; Levine, J.H.; Cogdill, A.P.; Zhao, Y.; Anang, N.A.S.; Andrews, M.C.; Sharma, P.; Wang, J.; Wargo, J.A.; Pe’er, D.; et al. Distinct Cellular Mechanisms Underlie Anti-CTLA-4 and Anti-PD-1 Checkpoint Blockade. Cell 2017, 170, 1120–1133. [Google Scholar] [CrossRef] [Green Version]
  108. Hellmann, M.D.; Rizvi, N.A.; Goldman, J.W.; Gettinger, S.N.; Borghaei, H.; Brahmer, J.R.; Ready, N.E.; Gerber, D.E.; Chow, L.Q.; Juergens, R.A.; et al. Nivolumab plus ipilimumab as first-line treatment for advanced non-small-cell lung cancer (CheckMate 012): Results of an open-label, phase 1, multicohort study. Lancet Oncol. 2017, 18, 31–41. [Google Scholar] [CrossRef] [Green Version]
  109. Antonia, S.; Goldberg, S.B.; Balmanoukian, A.; Chaft, J.E.; Sanborn, R.E.; Gupta, A.; Narwal, R.; Steele, K.; Gu, Y.; Karakunnel, J.J.; et al. Safety and antitumour activity of durvalumab plus tremelimumab in non-small cell lung cancer: A multicentre, phase 1b study. Lancet Oncol. 2016, 17, 299–308. [Google Scholar] [CrossRef] [Green Version]
  110. Disselhorst, M.J.; Quispel-Janssen, J.; Lalezari, F.; Monkhorst, K.; de Vries, J.F.; van der Noort, V.; Harms, E.; Burgers, S.; Baas, P. Ipilimumab and nivolumab in the treatment of recurrent malignant pleural mesothelioma (INITIATE): Results of a prospective, single-arm, phase 2 trial. Lancet Respir. Med. 2019, 7, 260–270. [Google Scholar] [CrossRef]
  111. Janjigian, Y.Y.; Bendell, J.; Calvo, E.; Kim, J.W.; Ascierto, P.A.; Sharma, P.; Ott, P.A.; Peltola, K.; Jaeger, D.; Evans, J.; et al. CheckMate-032 Study: Efficacy and Safety of Nivolumab and Nivolumab Plus Ipilimumab in Patients With Metastatic Esophagogastric Cancer. J. Clin. Oncol. 2018, 36, 2836–2844. [Google Scholar] [CrossRef] [PubMed]
  112. Boudadi, K.; Suzman, D.L.; Anagnostou, V.; Fu, W.; Luber, B.; Wang, H.; Niknafs, N.; White, J.R.; Silberstein, J.L.; Sullivan, R.; et al. Ipilimumab plus nivolumab and DNA-repair defects in AR-V7-expressing metastatic prostate cancer. Oncotarget 2018, 9, 28561–28571. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  113. D’Angelo, S.P.; Mahoney, M.R.; Van Tine, B.A.; Atkins, J.; Milhem, M.M.; Jahagirdar, B.N.; Antonescu, C.R.; Horvath, E.; Tap, W.D.; Schwartz, G.K.; et al. Nivolumab with or without ipilimumab treatment for metastatic sarcoma (Alliance A091401): Two open-label, non-comparative, randomised, phase 2 trials. Lancet Oncol. 2018, 19, 416–426. [Google Scholar] [CrossRef]
  114. Pol, J.; Vacchelli, E.; Aranda, F.; Castoldi, F.; Eggermont, A.; Cremer, I.; Sautès-Fridman, C.; Fucikova, J.; Galon, J.; Spisek, R.; et al. Trial Watch: Immunogenic cell death inducers for anticancer chemotherapy. Oncoimmunology 2015, 4, e1008866. [Google Scholar] [CrossRef] [PubMed]
  115. Jiang, W.; Chan, C.K.; Weissman, I.L.; Kim, B.Y.S.; Hahn, S.M. Immune Priming of the Tumor Microenvironment by Radiation. Trends Cancer 2016, 2, 638–645. [Google Scholar] [CrossRef] [PubMed]
  116. Wang, Y.J.; Fletcher, R.; Yu, J.; Zhang, L. Immunogenic effects of chemotherapy-induced tumor cell death. Genes Dis. 2018, 5, 194–203. [Google Scholar] [CrossRef]
  117. Tesniere, A.; Schlemmer, F.; Boige, V.; Kepp, O.; Martins, I.; Ghiringhelli, F.; Aymeric, L.; Michaud, M.; Apetoh, L.; Barault, L.; et al. Immunogenic death of colon cancer cells treated with oxaliplatin. Oncogene 2010, 29, 482–491. [Google Scholar] [CrossRef] [Green Version]
  118. Obeid, M.; Tesniere, A.; Ghiringhelli, F.; Fimia, G.M.; Apetoh, L.; Perfettini, J.L.; Castedo, M.; Mignot, G.; Panaretakis, T.; Casares, N.; et al. Calreticulin exposure dictates the immunogenicity of cancer cell death. Nat. Med. 2007, 13, 54–61. [Google Scholar] [CrossRef]
  119. Alizadeh, D.; Larmonier, N. Chemotherapeutic targeting of cancer-induced immunosuppressive cells. Cancer Res. 2014, 74, 2663–2668. [Google Scholar] [CrossRef] [Green Version]
  120. Pfirschke, C.; Engblom, C.; Rickelt, S.; Cortez-Retamozo, V.; Garris, C.; Pucci, F.; Yamazaki, T.; Poirier-Colame, V.; Newton, A.; Redouane, Y.; et al. Immunogenic Chemotherapy Sensitizes Tumors to Checkpoint Blockade Therapy. Immunity 2016, 44, 343–354. [Google Scholar] [CrossRef] [Green Version]
  121. Wang, Z.; Till, B.; Gao, Q. Chemotherapeutic agent-mediated elimination of myeloid-derived suppressor cells. Oncoimmunology 2017, 6, e1331807. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  122. Addeo, A.; Banna, G.L.; Metro, G.; Di Maio, M. Chemotherapy in Combination With Immune Checkpoint Inhibitors for the First-Line Treatment of Patients With Advanced Non-small Cell Lung Cancer: A Systematic Review and Literature-Based Meta-Analysis. Front. Oncol. 2019, 9, 264. [Google Scholar] [CrossRef] [Green Version]
  123. Lesterhuis, W.J.; Salmons, J.; Nowak, A.K.; Rozali, E.N.; Khong, A.; Dick, I.M.; Harken, J.A.; Robinson, B.W.; Lake, R.A. Synergistic effect of CTLA-4 blockade and cancer chemotherapy in the induction of anti-tumor immunity. PLoS ONE 2013, 8, e61895. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  124. Voorwerk, L.; Slagter, M.; Horlings, H.M.; Sikorska, K.; van de Vijver, K.K.; de Maaker, M.; Nederlof, I.; Kluin, R.J.C.; Warren, S.; Ong, S.; et al. Immune induction strategies in metastatic triple-negative breast cancer to enhance the sensitivity to PD-1 blockade: The TONIC trial. Nat. Med. 2019, 25, 920–928. [Google Scholar] [CrossRef] [PubMed]
  125. Reck, M.; Bondarenko, I.; Luft, A.; Serwatowski, P.; Barlesi, F.; Chacko, R.; Sebastian, M.; Lu, H.; Cuillerot, J.M.; Lynch, T.J. Ipilimumab in combination with paclitaxel and carboplatin as first-line therapy in extensive-disease-small-cell lung cancer: Results from a randomized, double-blind, multicenter phase 2 trial. Ann. Oncol. 2013, 24, 75–83. [Google Scholar] [CrossRef] [PubMed]
  126. Golden, E.B.; Frances, D.; Pellicciotta, I.; Demaria, S.; Helen Barcellos-Hoff, M.; Formenti, S.C. Radiation fosters dose-dependent and chemotherapy-induced immunogenic cell death. Oncoimmunology 2014, 3, e28518. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  127. Kachikwu, E.L.; Iwamoto, K.S.; Liao, Y.P.; DeMarco, J.J.; Agazaryan, N.; Economou, J.S.; McBride, W.H.; Schaue, D. Radiation enhances regulatory T cell representation. Int. J. Radiat. Oncol. Biol. Phys. 2011, 81, 1128–1135. [Google Scholar] [CrossRef] [Green Version]
  128. Barcellos-Hoff, M.H.; Derynck, R.; Tsang, M.L.; Weatherbee, J.A. Transforming growth factor-beta activation in irradiated murine mammary gland. J. Clin. Investig. 1994, 93, 892–899. [Google Scholar] [CrossRef]
  129. Demaria, S.; Kawashima, N.; Yang, A.M.; Devitt, M.L.; Babb, J.S.; Allison, J.P.; Formenti, S.C. Immune-mediated inhibition of metastases after treatment with local radiation and CTLA-4 blockade in a mouse model of breast cancer. Clin. Cancer Res. 2005, 11, 728–734. [Google Scholar]
  130. Zeng, J.; See, A.P.; Phallen, J.; Jackson, C.M.; Belcaid, Z.; Ruzevick, J.; Durham, N.; Meyer, C.; Harris, T.J.; Albesiano, E.; et al. Anti-PD-1 blockade and stereotactic radiation produce long-term survival in mice with intracranial gliomas. Int. J. Radiat. Oncol. Biol. Phys. 2013, 86, 343–349. [Google Scholar] [CrossRef] [Green Version]
  131. Kim, K.J.; Kim, J.H.; Lee, S.J.; Lee, E.J.; Shin, E.C.; Seong, J. Radiation improves antitumor effect of immune checkpoint inhibitor in murine hepatocellular carcinoma model. Oncotarget 2017, 8, 41242–41255. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  132. Wang, Y.; Deng, W.; Li, N.; Neri, S.; Sharma, A.; Jiang, W.; Lin, S.H. Combining Immunotherapy and Radiotherapy for Cancer Treatment: Current Challenges and Future Directions. Front. Pharmacol. 2018, 9, 185. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  133. Sharabi, A.B.; Nirschl, C.J.; Kochel, C.M.; Nirschl, T.R.; Francica, B.J.; Velarde, E.; Deweese, T.L.; Drake, C.G. Stereotactic Radiation Therapy Augments Antigen-Specific PD-1-Mediated Antitumor Immune Responses via Cross-Presentation of Tumor Antigen. Cancer Immunol. Res. 2015, 3, 345–355. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  134. Dovedi, S.J.; Adlard, A.L.; Lipowska-Bhalla, G.; McKenna, C.; Jones, S.; Cheadle, E.J.; Stratford, I.J.; Poon, E.; Morrow, M.; Stewart, R.; et al. Acquired resistance to fractionated radiotherapy can be overcome by concurrent PD-L1 blockade. Cancer Res. 2014, 74, 5458–5468. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  135. Deng, L.; Liang, H.; Burnette, B.; Beckett, M.; Darga, T.; Weichselbaum, R.R.; Fu, Y.X. Irradiation and anti-PD-L1 treatment synergistically promote antitumor immunity in mice. J. Clin. Investig. 2014, 124, 687–695. [Google Scholar] [CrossRef] [PubMed]
  136. Antonia, S.J.; Villegas, A.; Daniel, D.; Vicente, D.; Murakami, S.; Hui, R.; Yokoi, T.; Chiappori, A.; Lee, K.H.; de Wit, M.; et al. Durvalumab after Chemoradiotherapy in Stage III Non-Small-Cell Lung Cancer. N. Engl. J. Med. 2017, 377, 1919–1929. [Google Scholar] [CrossRef]
  137. Abuodeh, Y.; Venkat, P.; Kim, S. Systematic review of case reports on the abscopal effect. Curr. Probl. Cancer 2016, 40, 25–37. [Google Scholar] [CrossRef]
  138. Demaria, S.; Ng, B.; Devitt, M.L.; Babb, J.S.; Kawashima, N.; Liebes, L.; Formenti, S.C. Ionizing radiation inhibition of distant untreated tumors (abscopal effect) is immune mediated. Int. J. Radiat. Oncol. Biol. Phys. 2004, 58, 862–870. [Google Scholar] [CrossRef]
  139. Dewan, M.Z.; Galloway, A.E.; Kawashima, N.; Dewyngaert, J.K.; Babb, J.S.; Formenti, S.C.; Demaria, S. Fractionated but not single-dose radiotherapy induces an immune-mediated abscopal effect when combined with anti-CTLA-4 antibody. Clin. Cancer Res. 2009, 15, 5379–5388. [Google Scholar] [CrossRef]
  140. Park, S.S.; Dong, H.; Liu, X.; Harrington, S.M.; Krco, C.J.; Grams, M.P.; Mansfield, A.S.; Furutani, K.M.; Olivier, K.R.; Kwon, E.D. PD-1 Restrains Radiotherapy-Induced Abscopal Effect. Cancer Immunol. Res. 2015, 3, 610–619. [Google Scholar] [CrossRef] [Green Version]
  141. Postow, M.A.; Callahan, M.K.; Barker, C.A.; Yamada, Y.; Yuan, J.; Kitano, S.; Mu, Z.; Rasalan, T.; Adamow, M.; Ritter, E.; et al. Immunologic correlates of the abscopal effect in a patient with melanoma. N. Engl. J. Med. 2012, 366, 925–931. [Google Scholar] [CrossRef] [Green Version]
  142. Badalamenti, G.; Fanale, D.; Incorvaia, L.; Barraco, N.; Listì, A.; Maragliano, R.; Vincenzi, B.; Calò, V.; Iovanna, J.L.; Bazan, V.; et al. Role of tumor-infiltrating lymphocytes in patients with solid tumors: Can a drop dig a stone? Cell Immunol. 2019, 343. [Google Scholar] [CrossRef]
  143. Hiniker, S.M.; Reddy, S.A.; Maecker, H.T.; Subrahmanyam, P.B.; Rosenberg-Hasson, Y.; Swetter, S.M.; Saha, S.; Shura, L.; Knox, S.J. A Prospective Clinical Trial Combining Radiation Therapy With Systemic Immunotherapy in Metastatic Melanoma. Int. J. Radiat. Oncol. Biol. Phys. 2016, 96, 578–588. [Google Scholar] [CrossRef] [Green Version]
  144. Anderson, E.S.; Postow, M.A.; Wolchok, J.D.; Young, R.J.; Ballangrud, Å.; Chan, T.A.; Yamada, Y.; Beal, K. Melanoma brain metastases treated with stereotactic radiosurgery and concurrent pembrolizumab display marked regression; efficacy and safety of combined treatment. J. Immunother. Cancer 2017, 5, 76. [Google Scholar] [CrossRef] [Green Version]
  145. Shaverdian, N.; Lisberg, A.E.; Bornazyan, K.; Veruttipong, D.; Goldman, J.W.; Formenti, S.C.; Garon, E.B.; Lee, P. Previous radiotherapy and the clinical activity and toxicity of pembrolizumab in the treatment of non-small-cell lung cancer: A secondary analysis of the KEYNOTE-001 phase 1 trial. Lancet Oncol. 2017, 18, 895–903. [Google Scholar] [CrossRef]
  146. Yamaguchi, K.; Takagi, Y.; Aoki, S.; Futamura, M.; Saji, S. Significant detection of circulating cancer cells in the blood by reverse transcriptase-polymerase chain reaction during colorectal cancer resection. Ann. Surg. 2000, 232, 58–65. [Google Scholar] [CrossRef]
  147. Belizon, A.; Balik, E.; Feingold, D.L.; Bessler, M.; Arnell, T.D.; Forde, K.A.; Horst, P.K.; Jain, S.; Cekic, V.; Kirman, I.; et al. Major abdominal surgery increases plasma levels of vascular endothelial growth factor: Open more so than minimally invasive methods. Ann. Surg. 2006, 244, 792–798. [Google Scholar] [CrossRef]
  148. Tai, L.H.; de Souza, C.T.; Bélanger, S.; Ly, L.; Alkayyal, A.A.; Zhang, J.; Rintoul, J.L.; Ananth, A.A.; Lam, T.; Breitbach, C.J.; et al. Preventing postoperative metastatic disease by inhibiting surgery-induced dysfunction in natural killer cells. Cancer Res. 2013, 73, 97–107. [Google Scholar] [CrossRef] [Green Version]
  149. Yakar, I.; Melamed, R.; Shakhar, G.; Shakhar, K.; Rosenne, E.; Abudarham, N.; Page, G.G.; Ben-Eliyahu, S. Prostaglandin e(2) suppresses NK activity in vivo and promotes postoperative tumor metastasis in rats. Ann. Surg. Oncol. 2003, 10, 469–479. [Google Scholar] [CrossRef]
  150. Liu, J.; Blake, S.J.; Yong, M.C.; Harjunpää, H.; Ngiow, S.F.; Takeda, K.; Young, A.; O’Donnell, J.S.; Allen, S.; Smyth, M.J.; et al. Improved Efficacy of Neoadjuvant Compared to Adjuvant Immunotherapy to Eradicate Metastatic Disease. Cancer Discov. 2016, 6, 1382–1399. [Google Scholar] [CrossRef] [Green Version]
  151. Wang, C.; Sun, W.; Ye, Y.; Hu, Q.; Bomba, H.N.; Gu, Z. In situ activation of platelets with checkpoint inhibitors for post-surgical cancer immunotherapy. Nat. Biomed. Eng. 2017, 1. [Google Scholar] [CrossRef]
  152. Sun, Z.; Mao, A.; Wang, Y.; Zhao, Y.; Chen, J.; Xu, P.; Miao, C. Treatment with anti-programmed cell death 1 (PD-1) antibody restored postoperative CD8+ T cell dysfunction by surgical stress. Biomed. Pharmacother. 2017, 89, 1235–1241. [Google Scholar] [CrossRef] [PubMed]
  153. Forde, P.M.; Chaft, J.E.; Smith, K.N.; Anagnostou, V.; Cottrell, T.R.; Hellmann, M.D.; Zahurak, M.; Yang, S.C.; Jones, D.R.; Broderick, S.; et al. Neoadjuvant PD-1 Blockade in Resectable Lung Cancer. N. Engl. J. Med. 2018, 378, 1976–1986. [Google Scholar] [CrossRef] [PubMed]
  154. Carthon, B.C.; Wolchok, J.D.; Yuan, J.; Kamat, A.; Ng Tang, D.S.; Sun, J.; Ku, G.; Troncoso, P.; Logothetis, C.J.; Allison, J.P.; et al. Preoperative CTLA-4 blockade: Tolerability and immune monitoring in the setting of a presurgical clinical trial. Clin. Cancer Res. 2010, 16, 2861–2871. [Google Scholar] [CrossRef] [Green Version]
  155. Tarhini, A.A.; Edington, H.; Butterfield, L.H.; Lin, Y.; Shuai, Y.; Tawbi, H.; Sander, C.; Yin, Y.; Holtzman, M.; Johnson, J.; et al. Immune monitoring of the circulation and the tumor microenvironment in patients with regionally advanced melanoma receiving neoadjuvant ipilimumab. PLoS ONE 2014, 9, e87705. [Google Scholar] [CrossRef] [Green Version]
  156. Curran, M.A.; Glisson, B.S. New Hope for Therapeutic Cancer Vaccines in the Era of Immune Checkpoint Modulation. Annu Rev. Med. 2019, 70, 409–424. [Google Scholar] [CrossRef]
  157. Anassi, E.; Ndefo, U.A. Sipuleucel-T (provenge) injection: The first immunotherapy agent (vaccine) for hormone-refractory prostate cancer. Pharm. Ther. 2011, 36, 197–202. [Google Scholar]
  158. van Elsas, A.; Hurwitz, A.A.; Allison, J.P. Combination immunotherapy of B16 melanoma using anti-cytotoxic T lymphocyte-associated antigen 4 (CTLA-4) and granulocyte/macrophage colony-stimulating factor (GM-CSF)-producing vaccines induces rejection of subcutaneous and metastatic tumors accompanied by autoimmune depigmentation. J. Exp. Med. 1999, 190, 355–366. [Google Scholar] [CrossRef]
  159. Hurwitz, A.A.; Foster, B.A.; Kwon, E.D.; Truong, T.; Choi, E.M.; Greenberg, N.M.; Burg, M.B.; Allison, J.P. Combination immunotherapy of primary prostate cancer in a transgenic mouse model using CTLA-4 blockade. Cancer Res. 2000, 60, 2444–2448. [Google Scholar]
  160. Wada, S.; Jackson, C.M.; Yoshimura, K.; Yen, H.R.; Getnet, D.; Harris, T.J.; Goldberg, M.V.; Bruno, T.C.; Grosso, J.F.; Durham, N.; et al. Sequencing CTLA-4 blockade with cell-based immunotherapy for prostate cancer. J. Transl. Med. 2013, 11, 89. [Google Scholar] [CrossRef] [Green Version]
  161. Soares, K.C.; Rucki, A.A.; Wu, A.A.; Olino, K.; Xiao, Q.; Chai, Y.; Wamwea, A.; Bigelow, E.; Lutz, E.; Liu, L.; et al. PD-1/PD-L1 blockade together with vaccine therapy facilitates effector T-cell infiltration into pancreatic tumors. J. Immunother. 2015, 38, 1–11. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  162. Antonios, J.P.; Soto, H.; Everson, R.G.; Orpilla, J.; Moughon, D.; Shin, N.; Sedighim, S.; Yong, W.H.; Li, G.; Cloughesy, T.F.; et al. PD-1 blockade enhances the vaccination-induced immune response in glioma. JCI Insight 2016, 1. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  163. Le, D.T.; Lutz, E.; Uram, J.N.; Sugar, E.A.; Onners, B.; Solt, S.; Zheng, L.; Diaz, L.A.; Donehower, R.C.; Jaffee, E.M.; et al. Evaluation of ipilimumab in combination with allogeneic pancreatic tumor cells transfected with a GM-CSF gene in previously treated pancreatic cancer. J. Immunother. 2013, 36, 382–389. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  164. Massarelli, E.; William, W.; Johnson, F.; Kies, M.; Ferrarotto, R.; Guo, M.; Feng, L.; Lee, J.J.; Tran, H.; Kim, Y.U.; et al. Combining Immune Checkpoint Blockade and Tumor-Specific Vaccine for Patients With Incurable Human Papillomavirus 16-Related Cancer: A Phase 2 Clinical Trial. JAMA Oncol. 2019, 5, 67–73. [Google Scholar] [CrossRef] [Green Version]
  165. Chung, V.; Kos, F.J.; Hardwick, N.; Yuan, Y.; Chao, J.; Li, D.; Waisman, J.; Li, M.; Zurcher, K.; Frankel, P.; et al. Evaluation of safety and efficacy of p53MVA vaccine combined with pembrolizumab in patients with advanced solid cancers. Clin. Transl. Oncol. 2019, 21, 363–372. [Google Scholar] [CrossRef]
  166. June, C.H.; Riddell, S.R.; Schumacher, T.N. Adoptive cellular therapy: A race to the finish line. Sci. Transl. Med. 2015, 7, 280ps287. [Google Scholar] [CrossRef]
  167. Rosenberg, S.A.; Yang, J.C.; Sherry, R.M.; Kammula, U.S.; Hughes, M.S.; Phan, G.Q.; Citrin, D.E.; Restifo, N.P.; Robbins, P.F.; Wunderlich, J.R.; et al. Durable complete responses in heavily pretreated patients with metastatic melanoma using T-cell transfer immunotherapy. Clin. Cancer Res. 2011, 17, 4550–4557. [Google Scholar] [CrossRef] [Green Version]
  168. Maude, S.L.; Shpall, E.J.; Grupp, S.A. Chimeric antigen receptor T-cell therapy for ALL. Hematol. Am. Soc. Hematol. Educ. Program 2014, 2014, 559–564. [Google Scholar] [CrossRef] [Green Version]
  169. Lee, H.J.; Kim, Y.A.; Sim, C.K.; Heo, S.H.; Song, I.H.; Park, H.S.; Park, S.Y.; Bang, W.S.; Park, I.A.; Lee, M.; et al. Expansion of tumor-infiltrating lymphocytes and their potential for application as adoptive cell transfer therapy in human breast cancer. Oncotarget 2017, 8, 113345–113359. [Google Scholar] [CrossRef] [Green Version]
  170. Ben-Avi, R.; Farhi, R.; Ben-Nun, A.; Gorodner, M.; Greenberg, E.; Markel, G.; Schachter, J.; Itzhaki, O.; Besser, M.J. Establishment of adoptive cell therapy with tumor infiltrating lymphocytes for non-small cell lung cancer patients. Cancer Immunol. Immunother. 2018, 67, 1221–1230. [Google Scholar] [CrossRef]
  171. Klebanoff, C.A.; Rosenberg, S.A.; Restifo, N.P. Prospects for gene-engineered T cell immunotherapy for solid cancers. Nat. Med. 2016, 22, 26–36. [Google Scholar] [CrossRef] [Green Version]
  172. Kakarla, S.; Gottschalk, S. CAR T cells for solid tumors: Armed and ready to go? Cancer J. 2014, 20, 151–155. [Google Scholar] [CrossRef] [Green Version]
  173. Yoon, D.H.; Osborn, M.J.; Tolar, J.; Kim, C.J. Incorporation of Immune Checkpoint Blockade into Chimeric Antigen Receptor T Cells (CAR-Ts): Combination or Built-In CAR-T. Int. J. Mol. Sci. 2018, 19, 340. [Google Scholar] [CrossRef] [Green Version]
  174. Shi, L.Z.; Gao, J.; Allison, J.P.; Sharma, P. Combination therapy of adoptive T cell therapy and immune checkpoint blockades engages distinct mechanisms in CD4+ and CD8+ T cells. J. Immunol. 2018, 200, 122.21. [Google Scholar]
  175. Cao, Y.; Lu, W.; Sun, R.; Jin, X.; Cheng, L.; He, X.; Wang, L.; Yuan, T.; Lyu, C.; Zhao, M. Anti-CD19 Chimeric Antigen Receptor T Cells in Combination With Nivolumab Are Safe and Effective Against Relapsed/Refractory B-Cell Non-hodgkin Lymphoma. Front. Oncol. 2019, 9, 767. [Google Scholar] [CrossRef] [Green Version]
  176. John, L.B.; Devaud, C.; Duong, C.P.; Yong, C.S.; Beavis, P.A.; Haynes, N.M.; Chow, M.T.; Smyth, M.J.; Kershaw, M.H.; Darcy, P.K. Anti-PD-1 antibody therapy potently enhances the eradication of established tumors by gene-modified T cells. Clin. Cancer Res. 2013, 19, 5636–5646. [Google Scholar] [CrossRef] [Green Version]
  177. Cherkassky, L.; Morello, A.; Villena-Vargas, J.; Feng, Y.; Dimitrov, D.S.; Jones, D.R.; Sadelain, M.; Adusumilli, P.S. Human CAR T cells with cell-intrinsic PD-1 checkpoint blockade resist tumor-mediated inhibition. J. Clin. Investig. 2016, 126, 3130–3144. [Google Scholar] [CrossRef] [Green Version]
  178. Meyer, C.; Cagnon, L.; Costa-Nunes, C.M.; Baumgaertner, P.; Montandon, N.; Leyvraz, L.; Michielin, O.; Romano, E.; Speiser, D.E. Frequencies of circulating MDSC correlate with clinical outcome of melanoma patients treated with ipilimumab. Cancer Immunol. Immunother. 2014, 63, 247–257. [Google Scholar] [CrossRef] [Green Version]
  179. Bjoern, J.; Juul Nitschke, N.; Zeeberg Iversen, T.; Schmidt, H.; Fode, K.; Svane, I.M. Immunological correlates of treatment and response in stage IV malignant melanoma patients treated with Ipilimumab. Oncoimmunology 2016, 5, e1100788. [Google Scholar] [CrossRef] [Green Version]
  180. 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. [Google Scholar] [CrossRef] [Green Version]
  181. Genard, G.; Lucas, S.; Michiels, C. Reprogramming of Tumor-Associated Macrophages with Anticancer Therapies: Radiotherapy versus Chemo- and Immunotherapies. Front. Immunol. 2017, 8, 828. [Google Scholar] [CrossRef] [Green Version]
  182. 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]
  183. Dhupkar, P.; Gordon, N.; Stewart, J.; Kleinerman, E.S. Anti-PD-1 therapy redirects macrophages from an M2 to an M1 phenotype inducing regression of OS lung metastases. Cancer Med. 2018, 7, 2654–2664. [Google Scholar] [CrossRef]
  184. Orillion, A.; Hashimoto, A.; Damayanti, N.; Shen, L.; Adelaiye-Ogala, R.; Arisa, S.; Chintala, S.; Ordentlich, P.; Kao, C.; Elzey, B.; et al. Entinostat Neutralizes Myeloid-Derived Suppressor Cells and Enhances the Antitumor Effect of PD-1 Inhibition in Murine Models of Lung and Renal Cell Carcinoma. Clin. Cancer Res. 2017, 23, 5187–5201. [Google Scholar] [CrossRef] [Green Version]
  185. Zhu, Y.; Knolhoff, B.L.; Meyer, M.A.; Nywening, T.M.; West, B.L.; Luo, J.; Wang-Gillam, A.; Goedegebuure, S.P.; Linehan, D.C.; DeNardo, D.G. CSF1/CSF1R blockade reprograms tumor-infiltrating macrophages and improves response to T-cell checkpoint immunotherapy in pancreatic cancer models. Cancer Res. 2014, 74, 5057–5069. [Google Scholar] [CrossRef] [Green Version]
  186. McLean, G.W.; Carragher, N.O.; Avizienyte, E.; Evans, J.; Brunton, V.G.; Frame, M.C. The role of focal-adhesion kinase in cancer-a new therapeutic opportunity. Nat. Rev. Cancer 2005, 5, 505–515. [Google Scholar] [CrossRef]
  187. Stokes, J.B.; Adair, S.J.; Slack-Davis, J.K.; Walters, D.M.; Tilghman, R.W.; Hershey, E.D.; Lowrey, B.; Thomas, K.S.; Bouton, A.H.; Hwang, R.F.; et al. Inhibition of focal adhesion kinase by PF-562,271 inhibits the growth and metastasis of pancreatic cancer concomitant with altering the tumor microenvironment. Mol. Cancer Ther. 2011, 10, 2135–2145. [Google Scholar] [CrossRef] [Green Version]
  188. Serrels, A.; Lund, T.; Serrels, B.; Byron, A.; McPherson, R.C.; von Kriegsheim, A.; Gómez-Cuadrado, L.; Canel, M.; Muir, M.; Ring, J.E.; et al. Nuclear FAK controls chemokine transcription, Tregs, and evasion of anti-tumor immunity. Cell 2015, 163, 160–173. [Google Scholar] [CrossRef] [Green Version]
  189. Jiang, H.; Hegde, S.; Knolhoff, B.L.; Zhu, Y.; Herndon, J.M.; Meyer, M.A.; Nywening, T.M.; Hawkins, W.G.; Shapiro, I.M.; Weaver, D.T.; et al. Targeting focal adhesion kinase renders pancreatic cancers responsive to checkpoint immunotherapy. Nat. Med. 2016, 22, 851–860. [Google Scholar] [CrossRef]
  190. Coussens, L.M.; Zitvogel, L.; Palucka, A.K. Neutralizing tumor-promoting chronic inflammation: A magic bullet? Science 2013, 339, 286–291. [Google Scholar] [CrossRef] [Green Version]
  191. Zelenay, S.; van der Veen, A.G.; Böttcher, J.P.; Snelgrove, K.J.; Rogers, N.; Acton, S.E.; Chakravarty, P.; Girotti, M.R.; Marais, R.; Quezada, S.A.; et al. Cyclooxygenase-Dependent Tumor Growth through Evasion of Immunity. Cell 2015, 162, 1257–1270. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  192. Markosyan, N.; Chen, E.P.; Smyth, E.M. Targeting COX-2 abrogates mammary tumorigenesis: Breaking cancer-associated suppression of immunosurveillance. Oncoimmunology 2014, 3, e29287. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  193. Lizotte, P.H.; Hong, R.L.; Luster, T.A.; Cavanaugh, M.E.; Taus, L.J.; Wang, S.; Dhaneshwar, A.; Mayman, N.; Yang, A.; Kulkarni, M.; et al. A High-Throughput Immune-Oncology Screen Identifies EGFR Inhibitors as Potent Enhancers of Antigen-Specific Cytotoxic T-lymphocyte Tumor Cell Killing. Cancer Immunol. Res. 2018, 6, 1511–1523. [Google Scholar] [CrossRef] [Green Version]
  194. Tu, M.M.; Lee, F.Y.F.; Jones, R.T.; Kimball, A.K.; Saravia, E.; Graziano, R.F.; Coleman, B.; Menard, K.; Yan, J.; Michaud, E.; et al. Targeting DDR2 enhances tumor response to anti-PD-1 immunotherapy. Sci. Adv. 2019, 5. [Google Scholar] [CrossRef] [Green Version]
  195. Patel, S.J.; Sanjana, N.E.; Kishton, R.J.; Eidizadeh, A.; Vodnala, S.K.; Cam, M.; Gartner, J.J.; Jia, L.; Steinberg, S.M.; Yamamoto, T.N.; et al. Identification of essential genes for cancer immunotherapy. Nature 2017, 548, 537–542. [Google Scholar] [CrossRef] [Green Version]
  196. LaFleur, M.W.; Nguyen, T.H.; Coxe, M.A.; Yates, K.B.; Trombley, J.D.; Weiss, S.A.; Brown, F.D.; Gillis, J.E.; Coxe, D.J.; Doench, J.G.; et al. A CRISPR-Cas9 delivery system for in vivo screening of genes in the immune system. Nat. Commun. 2019, 10, 1668. [Google Scholar] [CrossRef] [Green Version]
  197. LaFleur, M.W.; Nguyen, T.H.; Coxe, M.A.; Miller, B.C.; Yates, K.B.; Gillis, J.E.; Sen, D.R.; Gaudiano, E.F.; Al Abosy, R.; Freeman, G.J.; et al. PTPN2 regulates the generation of exhausted CD8. Nat. Immunol. 2019, 20, 1335–1347. [Google Scholar] [CrossRef]

Share and Cite

MDPI and ACS Style

Tran, L.; Theodorescu, D. Determinants of Resistance to Checkpoint Inhibitors. Int. J. Mol. Sci. 2020, 21, 1594. https://doi.org/10.3390/ijms21051594

AMA Style

Tran L, Theodorescu D. Determinants of Resistance to Checkpoint Inhibitors. International Journal of Molecular Sciences. 2020; 21(5):1594. https://doi.org/10.3390/ijms21051594

Chicago/Turabian Style

Tran, Linda, and Dan Theodorescu. 2020. "Determinants of Resistance to Checkpoint Inhibitors" International Journal of Molecular Sciences 21, no. 5: 1594. https://doi.org/10.3390/ijms21051594

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