Next Article in Journal
Complications and Discharge after Radical Cystectomy for Older Patients with Muscle-Invasive Bladder Cancer: The ELCAPA-27 Cohort Study
Next Article in Special Issue
Preventing and Overcoming Resistance to PARP Inhibitors: A Focus on the Clinical Landscape
Previous Article in Journal
Comparison of Overall Survival between Surgical Resection and Radiofrequency Ablation for Hepatitis B-Related Hepatocellular Carcinoma
Previous Article in Special Issue
Structural Chromosome Instability: Types, Origins, Consequences, and Therapeutic Opportunities
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Impact of DNA Damage Response—Targeted Therapies on the Immune Response to Tumours

by
Nura Lutfi
1,
Miguel Alejandro Galindo-Campos
1 and
José Yélamos
1,2,*
1
Cancer Research Program, Hospital del Mar Medical Research Institute (IMIM), 08003 Barcelona, Spain
2
Immunology Unit, Department of Pathology, Hospital del Mar, 08003 Barcelona, Spain
*
Author to whom correspondence should be addressed.
Cancers 2021, 13(23), 6008; https://doi.org/10.3390/cancers13236008
Submission received: 29 October 2021 / Revised: 22 November 2021 / Accepted: 26 November 2021 / Published: 29 November 2021
(This article belongs to the Special Issue DNA Damage Response Targeting: Challenges and Opportunities)

Abstract

:

Simple Summary

Targeting tumour-specific defects in the DNA damage response (DDR) presents an opportunity for new therapeutic approaches to selectively kill cancer cells. Although the therapeutic rationale of DDR-targeted agents initially focused on their actions against tumour cells, these agents might also alter the crosstalk between tumour cells and the immune system. Here, we discuss recent data showing that DDR-targeted agents affect the antitumour immune response both through direct actions on the immune system components and through indirect effects on expression of different molecules and pathways in tumour cells that underpin the tumour cell–immune system.

Abstract

The DNA damage response (DDR) maintains the stability of a genome faced with genotoxic insults (exogenous or endogenous), and aberrations of the DDR are a hallmark of cancer cells. These cancer-specific DDR defects present new therapeutic opportunities, and different compounds that inhibit key components of DDR have been approved for clinical use or are in various stages of clinical trials. Although the therapeutic rationale of these DDR-targeted agents initially focused on their action against tumour cells themselves, these agents might also impact the crosstalk between tumour cells and the immune system, which can facilitate or impede tumour progression. In this review, we summarise recent data on how DDR-targeted agents can affect the interactions between tumour cells and the components of the immune system, both by acting directly on the immune cells themselves and by altering the expression of different molecules and pathways in tumour cells that are critical for their relationship with the immune system. Obtaining an in-depth understanding of the mechanisms behind how DDR-targeted therapies affect the immune system, and their crosstalk with tumour cells, may provide invaluable clues for the rational development of new therapeutic strategies in cancer.

1. Introduction

The genome is exposed to a multitude of genotoxic insults, both exogenous (e.g., mutagenic chemicals, ionising radiation) and endogenous (e.g., reactive oxygen species), that generate DNA damage. Moreover, endogenous DNA damage can lead to generation of recombination intermediates and arrested forks. To preserve genomic integrity, all eukaryotic cells have evolved mechanisms that sense DNA lesions, signal their presence and then promote their repair. Concomitant to DNA break repair, a rapid signalling cascade must be coordinated at the lesion site, which leads to activation of cytostatic and cytotoxic responses that limit the expansion of the damaged cells. Collectively, these mechanisms are referred to as the DNA damage response (DDR) [1]. Aberrations in the DDR result in genomic instability, which is a hallmark of cancer cells but at the same time opens up cancer treatment opportunities by targeting tumour-specific DDR defects to selectively kill cancer cells [2].
Poly(ADP-ribose) polymerases inhibitors (PARPi) that target both the PARP-1 and PARP-2 enzymes (e.g., olaparib, niraparib, rucaparib and talazoparib) have been the first DDR-targeting compounds approved for treatment of some tumours bearing homologous recombination (HR) DNA repair deficiencies [3], acting on the principle of synthetic lethality [4,5]. Other critical components of the DDR, including ataxia telangiectasia and Rad3-related (ATR), ataxia telangiectasia mutated (ATM), DNA-dependent protein kinase (DNA-PK), Checkpoint kinase 1 (CHK1), Checkpoint kinase 2 (CHK2) and Wee1 kinase (WEE1), are also promising as targets for fighting cancer [6,7]. Compounds that aim to inhibit these proteins are now at different phases of clinical development as either monotherapies or in combination with other therapies, mainly immunotherapies; it is expected that some of them will be approved for clinical use in the short term [6,8] (clinicaltrials.gov; accessed on 1 October 2021) (Figure 1).
These DDR-targeting agents have mainly been considered for their intrinsic actions towards tumour cells per se. However, these compounds will also affect the immune response to tumours, both by acting directly on the components of the immune system and by modifying components and signalling pathways in tumour cells that shape their interactions with the immune system. In this review, we summarise recent data on how DDR-targeted agents affect the antitumour immune response, focusing mainly on clinically approved compounds. Understanding how DDR-targeted therapies affect (i) the immune system and (ii) the crosstalk between the immune system and tumour cells may provide invaluable clues for designing new (and refining existing) therapeutic strategies in cancer.

2. Effects of DDR-Targeted Therapies on the Cell Components of the Immune System

The initial design of DDR-targeting agents to selectively kill tumour cells focused on their effects on tumour cells per se. However, systemic administration of these agents is likely to also impact the immune system, which may affect their therapeutic actions by facilitating or impeding tumour progression. This systemic effect on the immune system affects both the immune cells present in the tumour microenvironment (TME) and the peripheral immune cells that are required to drive effective antitumour immune responses [9,10,11].
Interestingly, in preclinical mouse models, treatment with the PARPi olaparib over a short course results in depletion of reticulocytes, B-cell progenitors and immature thymocytes, while longer treatment results in myelosuppression [12]. Moreover, patients treated with PARPi show a variety of adverse effects, including lymphopenia and other haematological toxicities [13,14].
Recently, we observed that olaparib treatment significant decreases tumour growth when breast cancer AT-3 cells, which are sensitive to olaparib, are implanted into an immunodeficient syngeneic mouse model but not if they are implanted into immunocompetent mice, suggesting that the antitumour effect of olaparib on the AT-3 cancer cells competes with the potentially negative pro-cancer effect of olaparib on the immune system [15]. However, other evidence suggests that DDR-targeted therapies may increase the antitumour immune response through several different mechanisms, such as by increasing antigenicity, increasing the gene instability of tumour cells, activating cytosolic immunity or modulating different components that may affect interactions between tumour and immune cells [16]. Together, these different aspects of the DDR effect on the immune system highlight the complex relationships between cancer and immune cells.
Immune cells in the TME include both those associated with immunological tolerance to tumours (immunosuppressive function) and immune effector cells against tumours (antitumour function). Understanding the impact of DDR-targeted agents in the delicate balance between these two different functional immune cell types will be critical for learning how to regulate tumour fate (Figure 2).

2.1. Effects of DDR-Targeted Therapies in Cells with Immunosuppressive Functions

The effects of DDR-targeted agents on the recruitment and function of cells with immunosuppressive functions—and therefore tumour progression-promoting functions—are beginning to be understood. The main cells with immunosuppressive functions are myeloid-derived suppressor cells (MDSCs), M2-macrophages and regulatory T cells (Tregs).
MDSCs represent a heterogeneous group of immature myeloid cells associated with immunosuppressive functions in the TME that contribute to tumour progression [17]. Accordingly, interfering with their recruitment to the TME and/or blocking their functional activity may improve the immune response to tumours. For instance, olaparib treatment suppresses MDSC migration to the TME [18]. The mechanisms underlying this effect appear to be based on olaparib downregulating the expression of stromal cell-derived factor 1-alpha (SDF1α), which is released to the TME by cancer-associated fibroblasts (CAF) and reduces CXCR4-mediated MDSC migration [18]. Interestingly, this olaparib-mediated decrease of infiltrating MDSCs in the TME improved the therapeutic efficacy of chimeric antigen receptor (CAR)-T cells in a mouse model of breast cancer [18]. Likewise, using the PARPi talazoparib encapsulated in a bilayer of a nano-liposome also decreased the levels of MDSCs, in both tumour and spleen in a mouse model of breast cancer [19]. Similarly, olaparib treatment reduced the granulocyte component of the MDSC population in a mouse model of breast cancer susceptibility genes 1 (BRCA1)-deficient ovarian cancer [20]. Strikingly, in a colon cancer model, moderate doses of olaparib only had a minor impact on the migration of MDSCs into the tumour, but inhibited the immunosuppressive activity of intratumour MDSCs and enhanced the response to anti-PD-1 therapy [21]. In addition to PARPi, treatment with a combination of the CHK1i SRA737 and the antimetabolite agent gemcitabine plus anti-PD-L1/anti-PD-1 also decreased MDSCs in a small cell lung cancer (SCLC) model [22].
Macrophages differentiate from circulating monocytes after extravasation to tissues and play different roles, including tissue homeostasis, host defence, antigen presentation to T cells and modulation of inflammatory responses [23]. A characteristic feature of macrophages is their extensive plasticity, which allows them to adapt their phenotype in response to environmental cues [24]. Tumour-associated macrophages (TAMs) are classified into two main groups, according to their antitumour (M1) or pro-tumour (M2) properties [23]. Of note, olaparib treatment in combination with radiotherapy results in macrophage activation towards a proinflammatory M1 phenotype in a colorectal tumour model [25]. However, in a mouse model of BRCA1-deficient, triple-negative breast cancer (TNBC), olaparib treatment had a dual effect on macrophages, with an antitumour effect due to inducing macrophage expression of co-stimulatory molecules (CD80) and activation markers (CD40), and a pro-tumour effect due to increasing the expression of immunosuppressive markers (e.g., programmed death ligand-1, PD-L1) and colony-stimulating factor 1 receptor (CSF1R) [26]. Combining olaparib treatment with macrophage-targeting therapy by CSF1R-blocking antibodies significantly enhances the antitumour immune response and increases survival in mice with BRCA-deficient TNBC tumours [26]. In addition, treatment with a combination of the CHK1i SRA737, gemcitabine and anti-PD-L1/anti-PD-1 increased M1 macrophages and decreased M2 macrophages in a SCLC model [22].
Tregs comprise a subpopulation of T lymphocytes with immunosuppressive functions and therefore impaired immune response to tumours [27]. Previous studies have determined that mice deficient for PARP-1 have increased levels of Tregs [28,29], which may have contributed at least in part to the acceleration of tumourigenesis in the absence of PARP-1 in a c-Myc-driven B cell lymphoma mouse model [30]. However, despite the critical role of Tregs in modulating the immune response, studies about the effects of DDR-targeted agents on Tregs are very limited and show varying results. For instance, olaparib treatment reduced the increase in Treg levels observed in a mouse model of sepsis [31]. Nevertheless, in a BRCA1-deficient mouse model of ovarian cancer, the percentage of the intratumour Treg cell population was not modified after olaparib treatment [20]. In contrast, tumour-infiltrating Tregs were increased in a mouse model of melanoma treated with the CHK1i SRA737 plus a subclinical dose of hydroxyurea [32]. A combination therapy of the ATRi ceralasertib and radiation decreased the number of tumour-infiltrating Tregs in mouse models of KRAS-mutant cancer [33], as well as in a mouse model of hepatocellular carcinoma [34]. Altogether, these findings show the need for further studies that evaluate Tregs in patients receiving DDR-targeted agents.

2.2. Effects of DDR-Targeted Therapies on Immune Cells with Antitumoural Effector Functions

CD8 cytotoxic T cells and CD4 Th1 cells are the most important contributors to the adaptive immune cellular host defence against tumours [35]. T cell recognition of tumour antigens requires that these are processed by antigen-presenting cells (APCs)—mainly dendritic cells (DCs)—that present them in the context of self-major histocompatibility complex (MHC) molecules to naïve T cells in order to prime them [36]. This antigenic presentation, together with co-stimulatory signals, results in CD8 and CD4 naïve T cells differentiating into effector cells with antitumour activity. The antitumour functions of CD8 T cells are primarily mediated by the secretion of perforin and granzyme [37], whereas those of CD4 T cells are primarily mediated by the secretion of various cytokines such as IL-2 and tumour necrosis factor (TNF) that enhance the CD8 T cell response and activate other effector cells, such as natural killer (NK) cells [38].
Of note, we have previously demonstrated that dual deficiency for PARP-1 and PARP-2 in T cells results in a significant decrease of both CD4 and CD8 peripheral T cells [29] and consequently impairs the T cell response to tumours [15]. Although it seems clear that PARPi will not achieve the degree of inhibition that can be achieved by genetic deletion of PARP-1 and PARP-2, these inhibitors can affect the T cell compartment, which could compromise the T cell immune response to tumours. In this sense, it would be of interest to evaluate the T cell compartment (effector CD8 and CD4 vs. naïve) in PARPi-treated patients.
Most pre-clinically reported effects of DDR-targeted therapies on antitumour effector T cells have been attributed to the effects of these inhibitors on the tumour cell itself. For instance, therapies can activate the cGAS/STING pathway, thereby indirectly modulating T cell responses by modifying the relationship between the tumour and T cells (see also below). Nevertheless, some data have been obtained on the direct effects of DDR-targeted agents on T cells in which the cGAS/STING pathway does not play a role (or the role has not yet been determined). For example, the PARPi talazoparib significantly increased the number of peritoneal CD8 T cells as well as their production of interferon-γ (IFN-γ) and TNF-α in a mouse model of ovarian cancer [39]. Moreover, the activity of cytotoxic CD8 T cells and NKT cells increased in a mouse model of melanoma treated with the CHK1i SRA737 plus subclinical dose of hydroxyurea, in a cGAS/STING-independent manner [32]. The CHK1i (SRA737) in combination with gemcitabine and anti-PD-L1/anti-PD-1 increased antitumor CD8 cytotoxic T cells in a SCLC model [22]. In mouse models of KRAS-mutant cancer, the ATRi ceralasertib attenuated radiation-induced CD8 T cell exhaustion and potentiated CD8 T cell activity [33].
DCs play a critical role in the immune response against tumour cells through their role in antigen presentation to T cells, yet the effects of DDR-targeted therapies on their function is largely unknown. Ding et al. (2018) found that olaparib treatment increased levels of co-stimulatory effects of CD80 and CD86 molecules as well as of MHC-class II molecules on DCs, suggesting increased costimulatory and antigen-presentation activity to T cells [20]. In contrast, another study showed that olaparib does not affect DC differentiation or function [40]. These data indicate the need for further studies to clarify the effects of DDR-targeted agents on DC function and the impact this has on the antitumour immune response.
NK cells are components of the innate immune system and have an important role in the antitumour response, based on several of their functions: (i) their cytolytic activity via the release of perforin and granzyme; (ii) inducing apoptosis in target cells via the production of TNF; (iii) inducing apoptosis via cell–cell contact and activation of the tumour necrosis factor-related apoptosis-inducing ligand (TRAIL) and Fas ligand (FASL) pathways [41,42]; and (iv) killing tumour cells coated with antibodies via antibody-dependent cell-mediated cytotoxicity (ADCC) [43]. Many of the effects induced by DDR-targeted agents on the antitumour activities of NK cells described so far seem to be due to modifications of tumour cells themselves, which affect their interactions with NK cells (see below). Indeed, whether DDR-targeted compounds have any direct effects on NK cells themselves is largely unknown. Of note, treatment with the PARPi talazoparib significantly increased the number of peritoneal NK cells as well as their production of IFN-γ and TNF-α in a mouse model of ovarian cancer [39].

3. DDR-Targeted Agents Modulate Pathways in Tumour Cells That Impact Their Relationships with Immune Cells

DDR-targeted agents modulate different molecules and pathways in tumour cells that are critical in their relationships with the immune system; in this way, they may either limit or favour tumourigenesis (Figure 3).

3.1. DDR-Targeted Agents Modulate Immune Responses via Activation of the cGAS/STING Pathway in Tumour Cells

Recent data have shown that PARPi promotes accumulation of cytosolic DNA [44,45], via PARP-1 trapping-induced DDR [46]. This often results in micronuclei that can arise from unresolved genomic instability, from either lagging chromosomes and/or chromatin bridges [47,48]. ATRi treatment further increases the numbers of PARPi-induced micronuclei in BRCA2-deficient cancer cells [49]. Upon breakdown of the micronuclear envelope [50], double-strand DNA (dsDNA) accumulates in the cytoplasm and is sensed by signalling pathways, such as by cyclic GMP-AMP (cGAMP) synthase (cGAS) [51]. cGAS activation produces cGAMP, which then activates stimulator of IFN genes (STING) and leads to the activation of TANK-binding kinase 1 (TBK1), IκB kinase (IKK) and NF-κB inducing kinase (NIK). Altogether, activation of these kinases results in the activation and nuclear translocation of IFN regulatory factor 3 (IRF3) and NF-κB, resulting in expression of type I IFN, interferon-stimulated genes (ISGs) and inflammatory cytokines [52]—further linking DDR with the immune system [6,53]. Mounting evidence suggests the chronic activation of cGAS/STING can paradoxically induce an immune suppressive TME that promotes tumour progression [52,54,55]. Accordingly, cGAS/STING activation pathway may exert either an antitumour or pro-tumour effect, depending on several factors, such as the stage of tumour progression and the tissue-specific context.
Through activation of the cGAS/STING pathway, PARPi appear to induce a CD8 T cell–dependent antitumour response. For instance, olaparib treatment induced a strong antitumour immune response in different pre-clinical tumour mouse models, including TNBC [56], SCLC [57] and BRCA1-deficient ovarian cancer [20], in a cGAS/STING-dependent manner. In the TNBC model, this effect is more pronounced in HR-deficient than in HR-proficient TNBC [56]. In addition, olaparib treatment leads to cGAS/STING-associated inflammatory response in BRCA2-deficient cells, which is further increased by ATRi [49,58]. Olaparib and rucaparib treatment modulate the immune response through the cGAS/STING pathway in ERCC1-defective, non-small cell lung cancer (NSCLC) and BRCA1-defective TNBC cells [44]. On the other hand, in an NSCLC mouse model, the PARPi niraparib plus radiation activated STING, resulting in antitumour immunity [59]. In addition, high concentrations of olaparib or talazoparib also activate STING in BRCA-proficient cells [21]. Recently, it has also been suggested that PARPi-mediated modulation of the immune response is dependent on the PARP trapping activity of PARPi [37].
Beyond PARPi, the impact of other DDR-targeted agents on the immune response to cancer mediated by the cGAS/STING pathway is beginning to be elucidated. For example, treatment with the CHK1i prexasertib increased the level of tumour-infiltrating T lymphocytes and synergises with anti-PD-L1 immunotherapy in a mouse model of SCLC [57]. Treatment with different ATRi results in innate immune and T cell activation mediated by cGAS-STING pathway in various mouse models of cancer, including those for prostate cancer [60], hepatocellular carcinoma [34] and ovarian cancer [61]. The combination of a WEE1i (adavosertib) and an ATRi (ceralasertib) also promotes accumulation of cytosolic dsDNA, which subsequently activates the STING pathway and induces the production of type I IFN and the recruitment and activation of CD8 T cells—thereby inducing antitumour immunity [62]. Similar induction of antitumour immunity through STING activation has been reported after treatment with ATMi [63,64]. In contrast, one study revealed that despite increased levels of cytoplasmic DNA, treatment with a WEE1i (adavosertib) and CHK1i (prexasertib, MK-8776 and PF-477736) fails to activate a type I IFN response [65].
Altogether, these pre-clinical studies suggest that DDR-targeted agents activate the immune system through STING activation, providing a rationale for combining these agents with immune response modulators such as immune checkpoint inhibitors (ICIs). However, it has recently been shown in a ovarian cancer model that tumour-intrinsic STING promotes resistance to dual ICI therapy via vascular endothelial growth factor A (VEGF-A) [55]. This highlights both the complexity of the interactions between tumour cells and the immune system as well as the importance of properly exploring these interactions. Given that STING activation can have both pro- and antitumour effects, further studies are needed to determine the synergy of these combinations in different situations.

3.2. Regulation of the Expression of Immune Checkpoint Interacting Molecules on Tumour Cells by DDR-Targeted Agents

A key way in which tumour cells can escape an immune response is by expressing immune checkpoint interacting molecules on their surface, including PD-L1 and PD-L2 [66]. Engagement of programmed death protein-1 (PD1), expressed on activated T lymphocytes [67], with their ligands PD-L1 or PD-L2, inhibits T cell activation [68,69] and results in failure of T cells to respond to tumours [70]. Accordingly, the blocking antibodies anti-PD-1, anti-PD-Ll and anti-PD-L2, which are defined as immune checkpoint inhibitors (ICI), have been developed to stop this engagement and allow proper T cell responses to tumours [71]. In addition to monoclonal antibodies, other strategies to block these interactions are being developed, including using soluble PD-1 receptors capable of binding and neutralizing both PD-L2 and PD-L1 [72] and small-molecule inhibitors targeting PD-1/PD-L1 signalling pathway [73].
PARPi upregulates expression of PD-L1 in different cancer cells, including breast [26,74], SCLC [57], NSCLC [16] and biliary tract cancer [75], and accordingly improve cancer-associated immunosuppression [74]. Different mechanisms have been proposed to explain the upregulation of PD-L1 by PARPi, such as: inhibition of glycogen synthase kinase 3β (GSK3β) [74], resulting in PD-L1 stabilization [76]; IFN-γ-induced PD-L1 expression [44]; and suppression of nucleophosmin (NPM1) interactions with PARP1, thereby enhancing the association of NPM1 at the PD-L1 promoter [77]. PD-L1 is also upregulated by other DDR-targeted agents besides PARPi. For instance, upregulation of PD-L1 was observed after the combination of a WEE1i (adavosertib) and an ATRi (ceralasertib) [62]. Although PD-L2 may also play a similar role as PD-L1, the effects of DDR-targeted agents on PD-L2 expression have not been explored so far.
The tumour evasion mechanism mediated by PD-L1 upregulation in response to DDR-targeted agents can be reverted by combining these agents with the aforementioned antibodies that block the interactions of PD-L1 with PD1 [22,56,60,74] or with agents that alter PD-L1 expression [78]. Numerous clinical trials have been initiated that combine DDR-targeted agents with ICIs [6,16,79]. The results of some of these phase II clinical trials (Table 1) are already beginning to emerge, with varied outcomes. For instance, combining durvalumab (an anti-PD-L1 antibody) with olaparib in patients with relapsed SCLC did not meet the pre-set bar for efficacy [14]. Similarly, olaparib plus durvalumab in recurrent ovarian cancer shows only a modest clinical response [80]. In contrast, in metastatic castration-resistant prostate cancer, the durvalumab/olaparib combination demonstrates clinical efficacy, particularly in patients with DDR abnormalities [81]. In germline BRCA1/2-mutated metastatic breast cancer, the durvalumab/olaparib combination shows promising antitumour activity and safety [82]. In addition, the triple combination of durvalumab, olaparib and paclitaxel shows superior efficacy compared to standard neoadjuvant chemotherapy in human epidermal growth factor receptor 2 (HER2)-negative breast cancer [83]. Similar results have been found for other combinations. For example, treatment of patients with advanced or metastatic TNBC with the PARPi niraparib plus the anti-PD1 antibody pembrolizumab has provided encouraging antitumour activity, with higher response rates in patients with tumour-promoting BRCA mutations [84]. In metastatic NSCLC, niraparib plus pembrolizumab also shows clinical activity [85]. In addition, the niraparib/pembrolizumab combination in patients with recurrent platinum-resistant ovarian carcinoma also shows promising antitumour activity [86].
Despite the aforementioned encouraging preliminary results from some clinical trials, we need to cautiously await the results of other ongoing clinical trials and evaluated whether addition of ICIs improves long-term clinical outcomes as compared with DDR-targeted agents monotherapy, to better evaluate their effectiveness [6,79]. Likewise, it is very important to advance in the identification of biomarkers that allow us to predict the clinical response to the combination of DDR-targeted agents and ICI [87].

3.3. Modulation of Other Ligands on Tumour Cells by DDR-Targeted Agents That Affect Their Interactions with Immune Cells

The impact of DDR-targeted agents in the modulation of other surface molecules on tumour cells that will play a role in their crosstalk with the immune system has been poorly explored. Two good examples of these kinds of molecules are death receptors and NKG2D ligands.
As mentioned above, NK cells can destroy tumour cells by inducing death receptor-mediated apoptosis upon engagement of TRAIL and FASL with their receptors, TRAIL receptor (TRAILR) and FAS, respectively, at tumour target cells [42]. Of note, PARPi treatment induces the expression of these death receptors on the surface of tumour cells, thereby sensitising these cells to death receptor-mediated apoptosis [88,89] (Figure 3).
NKG2D is an activating receptor expressed on the surface of different immune cells, including NK cells, which recognises ligands (NKG2DL) that are upregulated on tumour cells, resulting in NK cell-mediated recognition and cytolysis of target cells [90,91]. DNA damage induction in tumours cells upregulates NKG2DL for the co-stimulatory receptors NKG2D in a STING-dependent manner, thereby stimulating the cytotoxicity of NK cells [92]. Of note, the PARPi olaparib upregulates expression of NKG2DL on the surface of cells of a human acute myeloid leukaemia cell line [93]. However, doxorubicin-induced NKG2DL expression on multiple myeloma cells is abolished after treatment with ATMi and ATRi [94].
Further studies are required to define the different roles of DDR-targeted agents in modulating the expression of these receptors and their impact on the immune response to tumours. Interestingly, inhibition of WEE1 by adavosertib sensitises head and neck cancer cells to NK lysis by overcoming the resistance to granzyme B-induced cell death [95].

4. Concluding Remarks and Future Prospects

The approval of PARPi for clinical use in different tumour types has highlighted the usefulness of targeting DDR components for cancer treatment. Accordingly, numerous clinical trials are underway with agents targeting key components of DDR. Although the therapeutic rationale was initially based on their effect on tumour cells per se, recent developments have shown that these compounds can also modulate the interactions between tumour cells and their environment, and especially with components of the immune system. The effects of DDR-targeted agents on the immune system can be direct, acting on immune cells, or indirect, for instance by modifying components of tumour cells that are critical in their communication with components of the immune response. Modulation of the immune response by DDR-targeted agents can either promote the immune response to the tumour, or conversely block this response and promote tumour progression—as reviewed here, there is experimental data supporting both situations. This apparent dichotomy underscores the complexity of the interactions between tumour cells and the immune system, which can vary depending on numerous factors. Currently, there is considerable enthusiasm about the prospect of DDR-targeted agents in activating the immune response to tumours by targeting STING pathway activation. It is important to be cautious in this regard. Although the majority of pre-clinical studies using different DDR-targeted agents that have been published seem to support this hypothesis, how cGAS/STING activation affects tumour development is controversial, as it has been shown to either limit or favour tumourigenesis. Similarly, the combination of DDR-targeted agents with ICI is also attracting much attention, as indicated by the large number of clinical trials underway. Clearly, it is necessary to await the results of these long-term trials before drawing conclusions about the clinical potential of these combinations. In this regard, it may be very important to identify biomarkers that can predict clinical situations in which DDR-targeted agents and ICI combinations may be useful. Future work will need address the immunomodulatory roles of DDR-targeting agents, to help design and optimise novel therapeutics strategies in cancer.

Author Contributions

Conceptualization, J.Y.; Figures, N.L.; Table, M.A.G.-C.; N.L., M.A.G.-C. and J.Y. wrote the manuscript. All authors have read and agreed to the published version of the manuscript.

Funding

The Yélamos lab is funded by the Spanish Ministerio de Ciencia e Innovación (grant PID2020-112526RB-I00 funded by MCIN/AEI/10.13039/501100011033) and The Fundación Científica de la Asociación Española Contra el Cáncer (AECC) (grant PROYEI6018YÉLA).

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Data Availability Statement

Not applicable.

Acknowledgments

The authors thank C. Ampurdanés for her helpful discussions, and V.A. Raker for manuscript editing. Images were created with BioRender.com accessed on 27 October 2021.

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Lecona, E.; Fernandez-Capetillo, O. Targeting ATR in cancer. Nat. Rev. Cancer 2018, 18, 586–595. [Google Scholar] [CrossRef] [PubMed]
  2. Wang, C.; Tang, M.; Chen, Z.; Nie, L.; Li, S.; Xiong, Y.; Szymonowicz, K.A.; Park, J.-M.; Zhang, H.; Feng, X.; et al. Genetic vulnerabilities upon inhibition of DNA damage response. Nucleic Acids Res. 2021, 49, 8214–8231. [Google Scholar] [CrossRef]
  3. Slade, D. PARP and PARG inhibitors in cancer treatment. Genes Dev. 2020, 34, 360–394. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  4. Farmer, H.; McCabe, N.; Lord, C.J.; Tutt, A.N.J.; Johnson, D.A.; Richardson, T.B.; Santarosa, M.; Dillon, K.J.; Hickson, I.; Knights, C.; et al. Targeting the DNA repair defect in BRCA mutant cells as a therapeutic strategy. Nature 2005, 434, 917–921. [Google Scholar] [CrossRef] [PubMed]
  5. Bryant, H.E.; Schultz, N.; Thomas, H.D.; Parker, K.M.; Flower, D.; Lopez, E.; Kyle, S.; Meuth, M.; Curtin, N.J.; Helleday, T. Specific killing of BRCA2-deficient tumours with inhibitors of poly(ADP-ribose) polymerase. Nature 2005, 434, 913–917. [Google Scholar] [CrossRef] [PubMed]
  6. Pilger, D.; Seymour, L.W.; Jackson, S.P. Interfaces between cellular responses to DNA damage and cancer immunotherapy. Genes Dev. 2021, 35, 602–618. [Google Scholar] [CrossRef]
  7. Pilié, P.G.; Tang, C.; Mills, G.B.; Yap, T.A. State-of-the-art strategies for targeting the DNA damage response in cancer. Nat. Rev. Clin. Oncol. 2019, 16, 81–104. [Google Scholar] [CrossRef]
  8. Barnieh, F.M.; Loadman, P.M.; Falconer, R.A. Progress towards a clinically-successful ATR inhibitor for cancer therapy. Curr. Res. Pharmacol. Drug Discov. 2021, 2, 100017. [Google Scholar] [CrossRef]
  9. Hiam-Galvez, K.J.; Allen, B.M.; Spitzer, M.H. Systemic immunity in cancer. Nat. Rev. Cancer 2021, 21, 345–359. [Google Scholar] [CrossRef]
  10. Shaked, Y. The pro-tumorigenic host response to cancer therapies. Nat. Rev. Cancer 2019, 19, 667–685. [Google Scholar] [CrossRef]
  11. Axelrod, M.L.; Nixon, M.J.; Gonzalez-Ericsson, P.I.; Bergman, R.E.; Pilkinton, M.A.; McDonnell, W.J.; Sanchez, V.; Opalenik, S.R.; Loi, S.; Zhou, J.; et al. Changes in Peripheral and Local Tumor Immunity after Neoadjuvant Chemotherapy Reshape Clinical Outcomes in Patients with Breast Cancer. Clin. Cancer Res. 2020, 26, 5668–5681. [Google Scholar] [CrossRef] [PubMed]
  12. Xu, Z.; Vandenberg, C.J.; Lieschke, E.; di Rago, L.; Scott, C.L.; Majewski, I.J. CHK2 Inhibition Provides a Strategy to Suppress Hematologic Toxicity from PARP Inhibitors. Mol. Cancer Res. 2021, 19, 1350–1360. [Google Scholar] [CrossRef]
  13. LaFargue, C.J.; Molin, G.Z.D.; Sood, A.K.; Coleman, R.L. Exploring and comparing adverse events between PARP inhibitors. Lancet Oncol. 2019, 20, e15–e28. [Google Scholar] [CrossRef]
  14. Thomas, A.; Vilimas, R.; Trindade, C.; Erwin-Cohen, R.; Roper, N.; Xi, L.; Krishnasamy, V.; Levy, E.; Mammen, A.; Nichols, S.; et al. Durvalumab in Combination with Olaparib in Patients with Relapsed SCLC: Results from a Phase II Study. J. Thorac. Oncol. 2019, 14, 1447–1457. [Google Scholar] [CrossRef] [PubMed]
  15. Moreno-Lama, L.; Galindo-Campos, M.A.; Martínez, C.; Comerma, L.; Vazquez, I.; Vernet-Tomas, M.; Ampurdanés, C.; Lutfi, N.; Martin-Caballero, J.; Dantzer, F.; et al. Coordinated signals from PARP-1 and PARP-2 are required to establish a proper T cell immune response to breast tumors in mice. Oncogene 2020, 39, 2835–2843. [Google Scholar] [CrossRef] [PubMed]
  16. Chabanon, R.M.; Rouanne, M.; Lord, C.J.; Soria, J.-C.; Pasero, P.; Postel-Vinay, S. Targeting the DNA damage response in immuno-oncology: Developments and opportunities. Nat. Rev. Cancer 2021, 21, 701–717. [Google Scholar] [CrossRef]
  17. Talmadge, J.E.; Gabrilovich, D.I. History of myeloid-derived suppressor cells. Nat. Rev. Cancer 2013, 13, 739–752. [Google Scholar] [CrossRef] [PubMed]
  18. Sun, R.; Luo, H.; Su, J.; Di, S.; Zhou, M.; Shi, B.; Sun, Y.; Du, G.; Zhang, H.; Jiang, H.; et al. Olaparib Suppresses MDSC Recruitment via SDF1α/CXCR4 Axis to Improve the Anti-tumor Efficacy of CAR-T Cells on Breast Cancer in Mice. Mol. Ther. 2021, 29, 60–74. [Google Scholar] [CrossRef] [PubMed]
  19. Zhang, D.; Baldwin, P.; Leal, A.S.M.; Carapellucci, S.; Sridhar, S.; Liby, K.T. A nano-liposome formulation of the PARP inhibitor Talazoparib enhances treatment efficacy and modulates immune cell populations in mammary tumors of BRCA-deficient mice. Theranostics 2019, 9, 6224–6238. [Google Scholar] [CrossRef] [PubMed]
  20. Ding, L.; Kim, H.-J.; Wang, Q.; Kearns, M.; Jiang, T.; Ohlson, C.E.; Li, B.B.; Xie, S.; Liu, J.F.; Stover, E.H.; et al. PARP Inhibition Elicits STING-Dependent Antitumor Immunity in Brca1-Deficient Ovarian Cancer. Cell Rep. 2018, 25, 2972–2980. [Google Scholar] [CrossRef] [Green Version]
  21. Ghonim, M.A.; Ibba, S.V.; Tarhuni, A.F.; Errami, Y.; Luu, H.H.; Dean, M.J.; El-Bahrawy, A.H.; Wyczechowska, D.; Benslimane, I.A.; del Valle, L.; et al. Targeting PARP-1 with metronomic therapy modulates MDSC suppressive function and enhances anti-PD-1 immunotherapy in colon cancer. J. Immunother. Cancer 2021, 9, e001643. [Google Scholar] [CrossRef]
  22. Sen, T.; della Corte, C.M.; Milutinovic, S.; Cardnell, R.J.; Diao, L.; Ramkumar, K.; Gay, C.M.; Stewart, C.A.; Fan, Y.; Shen, L.; et al. Combination Treatment of the Oral CHK1 Inhibitor, SRA737, and Low-Dose Gemcitabine Enhances the Effect of Programmed Death Ligand 1 Blockade by Modulating the Immune Microenvironment in SCLC. J. Thorac. Oncol. 2019, 14, 2152–2163. [Google Scholar] [CrossRef] [PubMed]
  23. Mehta, A.K.; Kadel, S.; Townsend, M.G.; Oliwa, M.; Guerriero, J.L. Macrophage Biology and Mechanisms of Immune Suppression in Breast Cancer. Front. Immunol. 2021, 12, 643771. [Google Scholar] [CrossRef]
  24. Gordon, S.; Plüddemann, A.; Martinez-Estrada, F. Macrophage heterogeneity in tissues: Phenotypic diversity and functions. Immunol. Rev. 2014, 262, 36–55. [Google Scholar] [CrossRef] [Green Version]
  25. Wu, Q.; Allouch, A.; Paoletti, A.; Leteur, C.; Mirjolet, C.; Martins, I.; Voisin, L.; Law, F.; Dakhli, H.; Mintet, E.; et al. NOX2-dependent ATM kinase activation dictates pro-inflammatory macrophage phenotype and improves effectiveness to radiation therapy. Cell Death Differ. 2017, 24, 1632–1644. [Google Scholar] [CrossRef]
  26. Mehta, A.K.; Cheney, E.M.; Hartl, C.A.; Pantelidou, C.; Oliwa, M.; Castrillon, J.A.; Lin, J.-R.; Hurst, K.E.; de Oliveira Taveira, M.; Johnson, N.T.; et al. Targeting immunosuppressive macrophages overcomes PARP inhibitor resistance in BRCA1-associated triple-negative breast cancer. Nat. Cancer 2021, 2, 66–82. [Google Scholar] [CrossRef]
  27. Glasner, A.; Plitas, G. Tumor resident regulatory T cells. Semin. Immunol. 2021, 52, 101476. [Google Scholar] [CrossRef] [PubMed]
  28. Nasta, F.; Laudisi, F.; Sambucci, M.; Rosado, M.M.; Pioli, C. Increased Foxp3+ Regulatory T Cells in Poly(ADP-Ribose) Polymerase-1 Deficiency. J. Immunol. 2010, 184, 3470–3477. [Google Scholar] [CrossRef]
  29. Navarro, J.; Gozalbo-López, B.; Méndez, A.C.; Dantzer, F.; Schreiber, V.; Martínez, C.; Arana, D.M.; Farrés, J.; Revilla-Nuin, B.; Bueno, M.F.; et al. PARP-1/PARP-2 double deficiency in mouse T cells results in faulty immune responses and T lymphomas. Sci. Rep. 2017, 7, 41962. [Google Scholar] [CrossRef] [Green Version]
  30. Galindo-Campos, M.A.; Lutfi, N.; Bonnin, S.; Martínez, C.; Velasco-Hernandez, T.; García-Hernández, V.; Martin-Caballero, J.; Ampurdanés, C.; Gimeno, R.; Colomo, L.; et al. Distinct roles for PARP-1 and PARP-2 in c-Myc-driven B-cell lymphoma in mice. Blood 2021. [Google Scholar] [CrossRef]
  31. Ahmad, A.; de Camargo Vieira, J.; de Mello, A.H.; de Lima, T.M.; Ariga, S.K.; Barbeiro, D.F.; Barbeiro, H.V.; Szczesny, B.; Törö, G.; Druzhyna, N.; et al. The PARP inhibitor olaparib exerts beneficial effects in mice subjected to cecal ligature and puncture and in cells subjected to oxidative stress without impairing DNA integrity: A potential opportunity for repurposing a clinically used oncological drug for the experimental therapy of sepsis. Pharmacol. Res. 2019, 145, 104263. [Google Scholar] [CrossRef] [Green Version]
  32. Proctor, M.; Gonzalez Cruz, J.L.; Daignault-Mill, S.M.; Veitch, M.; Zeng, B.; Ehmann, A.; Sabdia, M.; Snell, C.; Keane, C.; Dolcetti, R.; et al. Targeting Replication Stress Using CHK1 Inhibitor Promotes Innate and NKT Cell Immune Responses and Tumour Regression. Cancers 2021, 13, 3733. [Google Scholar] [CrossRef]
  33. Vendetti, F.P.; Karukonda, P.; Clump, D.A.; Teo, T.; LaLonde, R.; Nugent, K.; Ballew, M.; Kiesel, B.F.; Beumer, J.H.; Sarkar, S.N.; et al. ATR kinase inhibitor AZD6738 potentiates CD8+ T cell–dependent antitumor activity following radiation. J. Clin. Investig. 2018, 128, 3926–3940. [Google Scholar] [CrossRef]
  34. Sheng, H.; Huang, Y.; Xiao, Y.; Zhu, Z.; Shen, M.; Zhou, P.; Guo, Z.; Wang, J.; Wang, H.; Dai, W.; et al. ATR inhibitor AZD6738 enhances the antitumor activity of radiotherapy and immune checkpoint inhibitors by potentiating the tumor immune microenvironment in hepatocellular carcinoma. J. Immunother. Cancer 2020, 8, e000340. [Google Scholar] [CrossRef]
  35. Baumeister, S.H.; Freeman, G.J.; Dranoff, G.; Sharpe, A.H. Coinhibitory Pathways in Immunotherapy for Cancer. Annu. Rev. Immunol. 2016, 34, 539–573. [Google Scholar] [CrossRef]
  36. Wculek, S.K.; Cueto, F.J.; Mujal, A.M.; Melero, I.; Krummel, M.F.; Sancho, D. Dendritic cells in cancer immunology and immunotherapy. Nat. Rev. Immunol. 2020, 20, 7–24. [Google Scholar] [CrossRef]
  37. Matsushita, H.; Vesely, M.D.; Koboldt, D.C.; Rickert, C.G.; Uppaluri, R.; Magrini, V.J.; Arthur, C.D.; White, J.M.; Chen, Y.S.; Shea, L.K.; et al. Cancer exome analysis reveals a T-cell-dependent mechanism of cancer immunoediting. Nature 2012, 482, 400–404. [Google Scholar] [CrossRef]
  38. Pardoll, D.M.; Topalian, S.L. The role of CD4+ T cell responses in antitumor immunity. Curr. Opin. Immunol. 1998, 10, 588–594. [Google Scholar] [CrossRef]
  39. Huang, J.; Wang, L.; Cong, Z.; Amoozgar, Z.; Kiner, E.; Xing, D.; Orsulic, S.; Matulonis, U.; Goldberg, M.S. The PARP1 inhibitor BMN 673 exhibits immunoregulatory effects in a Brca1(-/-) murine model of ovarian cancer. Biochem. Biophys. Res. Commun. 2015, 463, 551–556. [Google Scholar] [CrossRef]
  40. Echeverria Tirado, L.C.; Ghonim, M.A.; Wang, J.; Al-Khami, A.A.; Wyczechowska, D.; Luu, H.H.; Kim, H.; Sanchez-Pino, M.D.; Yélamos, J.; Yassin, L.M.; et al. PARP-1 Is Critical for Recruitment of Dendritic Cells to the Lung in a Mouse Model of Asthma but Dispensable for Their Differentiation and Function. Mediat. Inflamm. 2019, 2019, 1656484. [Google Scholar] [CrossRef] [Green Version]
  41. Cerwenka, A.; Lanier, L.L. Natural killer cell memory in infection, inflammation and cancer. Nat. Rev. Immunol. 2016, 16, 112–123. [Google Scholar] [CrossRef]
  42. Prager, I.; Watzl, C. Mechanisms of natural killer cell-mediated cellular cytotoxicity. J. Leukoc. Biol. 2019, 105, 1319–1329. [Google Scholar] [CrossRef]
  43. Muntasell, A.; Rojo, F.; Servitja, S.; Rubio-Perez, C.; Cabo, M.; Tamborero, D.; Costa-Garcia, M.; Martínez-Garcia, M.; Menéndez, S.; Vazquez, I.; et al. NK Cell Infiltrates and HLA Class I Expression in Primary HER2+ Breast Cancer Predict and Uncouple Pathological Response and Disease-free Survival. Clin. Cancer Res. 2019, 25, 1535–1545. [Google Scholar] [CrossRef] [Green Version]
  44. Chabanon, R.M.; Muirhead, G.; Krastev, D.B.; Adam, J.; Morel, D.; Garrido, M.; Lamb, A.; Hénon, C.; Dorvault, N.; Rouanne, M.; et al. PARP inhibition enhances tumor cell–intrinsic immunity in ERCC1-deficient non–small cell lung cancer. J. Clin. Investig. 2019, 129, 1211–1228. [Google Scholar] [CrossRef]
  45. Shen, J.; Zhao, W.; Ju, Z.; Wang, L.; Peng, Y.; Labrie, M.; Yap, T.A.; Mills, G.B.; Peng, G. PARPi Triggers the STING-Dependent Immune Response and Enhances the Therapeutic Efficacy of Immune Checkpoint Blockade Independent of BRCAness. Cancer Res. 2019, 79, 311–319. [Google Scholar] [CrossRef] [Green Version]
  46. Kim, C.; Wang, X.-D.; Yu, Y. PARP1 inhibitors trigger innate immunity via PARP1 trapping-induced DNA damage response. eLife 2020, 9, 1–47. [Google Scholar] [CrossRef]
  47. MacKenzie, K.J.; Carroll, P.; Martin, C.-A.; Murina, O.; Fluteau, A.; Simpson, D.J.; Olova, N.; Sutcliffe, H.; Rainger, J.K.; Leitch, A.; et al. cGAS surveillance of micronuclei links genome instability to innate immunity. Nat. Cell Biol. 2017, 548, 461–465. [Google Scholar] [CrossRef] [Green Version]
  48. Sheng, J.; Chen, W.; Zhu, H.-J. The immune suppressive function of transforming growth factor-β(TGF-β) in human diseases. Growth Factors 2014, 33, 92–101. [Google Scholar] [CrossRef]
  49. Schoonen, P.M.; Kok, Y.P.; Wierenga, E.; Bakker, B.; Foijer, F.; Spierings, D.C.J.; van Vugt, M.A.T.M. Premature mitotic entry induced by ATR inhibition potentiates olaparib inhibition-mediated genomic instability, inflammatory signaling, and cytotoxicity in BRCA2-deficient cancer cells. Mol. Oncol. 2019, 13, 2422–2440. [Google Scholar] [CrossRef] [Green Version]
  50. Zhang, C.-Z.; Spektor, A.; Cornils, H.; Francis, J.M.; Jackson, E.K.; Liu, S.; Meyerson, M.; Pellman, D. Chromothripsis from DNA damage in micronuclei. Nat. Cell Biol. 2015, 522, 179–184. [Google Scholar] [CrossRef] [Green Version]
  51. Civril, F.; Deimling, T.; de Oliveira Mann, C.C.; Ablasser, A.; Moldt, M.; Witte, G.; Hornung, V.; Hopfner, K.-P. Structural mechanism of cytosolic DNA sensing by cGAS. Nature 2013, 498, 332–337. [Google Scholar] [CrossRef] [Green Version]
  52. Ahn, J.; Xia, T.; Konno, H.; Konno, K.; Ruiz, P.; Barber, G.N. Inflammation-driven carcinogenesis is mediated through STING. Nat. Commun. 2014, 5, 5166. [Google Scholar] [CrossRef] [Green Version]
  53. Paludan, S.R.; Reinert, L.S.; Hornung, V. DNA-stimulated cell death: Implications for host defence, inflammatory diseases and cancer. Nat. Rev. Immunol. 2019, 19, 141–153. [Google Scholar] [CrossRef]
  54. Kwon, J.; Bakhoum, S.F. The Cytosolic DNA-Sensing cGAS–STING Pathway in Cancer. Cancer Discov. 2020, 10, 26–39. [Google Scholar] [CrossRef]
  55. Bruand, M.; Barras, D.; Mina, M.; Ghisoni, E.; Morotti, M.; Lanitis, E.; Fahr, N.; Desbuisson, M.; Grimm, A.; Zhang, H.; et al. Cell-autonomous inflammation of BRCA1-deficient ovarian cancers drives both tumor-intrinsic immunoreactivity and immune resistance via STING. Cell Rep. 2021, 36, 109412. [Google Scholar] [CrossRef]
  56. Pantelidou, C.; Sonzogni, O.; de Oliveria Taveira, M.; Mehta, A.K.; Kothari, A.; Wang, D.; Visal, T.; Li, M.K.; Pinto, J.; Castrillon, J.A.; et al. PARP Inhibitor Efficacy Depends on CD8+ T-cell Recruitment via Intratumoral STING Pathway Activation in BRCA-Deficient Models of Triple-Negative Breast Cancer. Cancer Discov. 2019, 9, 722–737. [Google Scholar] [CrossRef] [Green Version]
  57. Sen, T.; Rodriguez, B.L.; Chen, L.; della Corte, C.M.; Morikawa, N.; Fujimoto, J.; Cristea, S.; Nguyen, T.; Diao, L.; Li, L.; et al. Targeting DNA Damage Response Promotes Antitumor Immunity through STING-Mediated T-cell Activation in Small Cell Lung Cancer. Cancer Discov. 2019, 9, 646–661. [Google Scholar] [CrossRef] [Green Version]
  58. Reisländer, T.; Lombardi, E.P.; Groelly, F.J.; Miar, A.; Porru, M.; di Vito, S.; Wright, B.; Lockstone, H.; Biroccio, A.; Harris, A.; et al. BRCA2 abrogation triggers innate immune responses potentiated by treatment with PARP inhibitors. Nat. Commun. 2019, 10, 3143. [Google Scholar] [CrossRef] [Green Version]
  59. Zhang, N.; Gao, Y.; Zeng, Z.; Luo, Y.; Jiang, X.; Zhang, J.; Li, J.; Gong, Y.; Xie, C. PARP inhibitor niraparib as a radiosensitizer promotes antitumor immunity of radiotherapy in EGFR-mutated non-small cell lung cancer. Clin. Transl. Oncol. 2021, 23, 1–11. [Google Scholar] [CrossRef]
  60. Tang, Z.; Pilié, P.G.; Geng, C.; Manyam, G.C.; Yang, G.; Park, S.; Wang, D.; Peng, S.; Wu, C.; Peng, G.; et al. ATR Inhibition Induces CDK1–SPOP Signaling and Enhances Anti–PD-L1 Cytotoxicity in Prostate Cancer. Clin. Cancer Res. 2021, 27, 4898–4909. [Google Scholar] [CrossRef]
  61. Chabanon, R.M.; Morel, D.; Eychenne, T.; Colmet-Daage, L.; Bajrami, I.; Dorvault, N.; Garrido, M.; Meisenberg, C.; Lamb, A.; Ngo, C.; et al. PBRM1 Deficiency Confers Synthetic Lethality to DNA Repair Inhibitors in Cancer. Cancer Res. 2021, 81, 2888–2902. [Google Scholar] [CrossRef]
  62. Wu, X.; Kang, X.; Zhang, X.; Xie, W.; Su, Y.; Liu, X.; Guo, L.; Guo, E.; Li, F.; Hu, D.; et al. WEE1 inhibitor and ataxia telangiectasia and RAD3-related inhibitor trigger stimulator of interferon gene-dependent immune response and enhance tumor treatment efficacy through programmed death-ligand 1 blockade. Cancer Sci. 2021, 112, 4444–4456. [Google Scholar] [CrossRef]
  63. Wang, L.; Yang, L.; Wang, C.; Zhao, W.; Ju, Z.; Zhang, W.; Shen, J.; Peng, Y.; An, C.; Luu, Y.T.; et al. Inhibition of the ATM/Chk2 axis promotes cGAS/STING signaling in ARID1A-deficient tumors. J. Clin. Investig. 2020, 130, 5951–5966. [Google Scholar] [CrossRef]
  64. Hu, M.; Zhou, M.; Bao, X.; Pan, D.; Jiao, M.; Liu, X.; Li, F.; Li, C.-Y. ATM inhibition enhances cancer immunotherapy by promoting mtDNA leakage and cGAS/STING activation. J. Clin. Investig. 2021, 131, e139333. [Google Scholar] [CrossRef]
  65. Wayne, J.; Brooks, T.; Landras, A.; Massey, A.J. Targeting DNA damage response pathways to activate the STING innate immune signaling pathway in human cancer cells. FEBS J. 2021, 288, 4507–4540. [Google Scholar] [CrossRef]
  66. Rabinovich, G.A.; Gabrilovich, D.; Sotomayor, E.M. Immunosuppressive Strategies that are Mediated by Tumor Cells. Annu. Rev. Immunol. 2007, 25, 267–296. [Google Scholar] [CrossRef] [Green Version]
  67. Agata, Y.; Kawasaki, A.; Nishimura, H.; Ishida, Y.; Tsubat, T.; Yagita, H.; Honjo, T. Expression of the PD-1 antigen on the surface of stimulated mouse T and B lymphocytes. Int. Immunol. 1996, 8, 765–772. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  68. 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]
  69. Freeman, G.J.; Long, A.J.; Iwai, Y.; Bourque, K.; Chernova, T.; Nishimura, H.N.; Fitz, L.J.; Malenkovich, N.; Okazaki, T.; Byrne, M.C.; et al. Engagement of the Pd-1 Immunoinhibitory Receptor by a Novel B7 Family Member Leads to Negative Regulation of Lymphocyte Activation. J. Exp. Med. 2000, 192, 1027–1034. [Google Scholar] [CrossRef] [Green Version]
  70. Blank, C.; Gajewski, T.F.; Mackensen, A. Interaction of PD-L1 on tumor cells with PD-1 on tumor-specific T cells as a mechanism of immune evasion: Implications for tumor immunotherapy. Cancer Immunol. Immunother. 2005, 54, 307–314. [Google Scholar] [CrossRef]
  71. Patsoukis, N.; Wang, Q.; Strauss, L.; Boussiotis, V.A. Revisiting the PD-1 pathway. Sci. Adv. 2020, 6, eabd2712. [Google Scholar] [CrossRef]
  72. Miao, Y.R.; Thakkar, K.N.; Qian, J.; Kariolis, M.S.; Huang, W.; Nandagopal, S.; Yang, T.T.C.; Diep, A.N.; Cherf, G.M.; Xu, Y.; et al. Neutralization of PD-L2 is Essential for Overcoming Immune Checkpoint Blockade Resistance in Ovarian Cancer. Clin. Cancer Res. 2021, 27, 4435–4448. [Google Scholar] [CrossRef]
  73. Awadasseid, A.; Wu, Y.; Zhang, W. Advance investigation on synthetic small-molecule inhibitors targeting PD-1/PD-L1 signaling pathway. Life Sci. 2021, 282, 119813. [Google Scholar] [CrossRef]
  74. Jiao, S.; Xia, W.; Yamaguchi, H.; Wei, Y.; Chen, M.-K.; Hsu, J.-M.; Hsu, J.L.; Yu, W.-H.; Du, Y.; Lee, H.-H.; et al. PARP Inhibitor Upregulates PD-L1 Expression and Enhances Cancer-Associated Immunosuppression. Clin. Cancer Res. 2017, 23, 3711–3720. [Google Scholar] [CrossRef] [Green Version]
  75. Nam, A.-R.; Yoon, J.; Jin, M.-H.; Bang, J.-H.; Oh, K.-S.; Seo, H.-R.; Kim, J.-M.; Kim, T.-Y.; Oh, D.-Y. ATR inhibition amplifies antitumor effects of olaparib in biliary tract cancer. Cancer Lett. 2021, 516, 38–47. [Google Scholar] [CrossRef]
  76. Li, C.-W.; Lim, S.-O.; Xia, W.; Lee, H.-H.; Chan, L.-C.; Kuo, C.-W.; Khoo, K.-H.; Chang, S.-S.; Cha, J.-H.; Kim, T.; et al. Glycosylation and stabilization of programmed death ligand-1 suppresses T-cell activity. Nat. Commun. 2016, 7, 12632. [Google Scholar] [CrossRef] [Green Version]
  77. Qin, G.; Wang, X.; Ye, S.; Li, Y.; Chen, M.; Wang, S.; Qin, T.; Zhang, C.; Li, Y.; Long, Q.; et al. NPM1 upregulates the transcription of PD-L1 and suppresses T cell activity in triple-negative breast cancer. Nat. Commun. 2020, 11, 1669. [Google Scholar] [CrossRef] [Green Version]
  78. Shao, B.; Li, C.-W.; Lim, S.-O.; Sun, L.; Lai, Y.-J.; Hou, J.; Liu, C.; Chang, C.-W.; Qiu, Y.; Hsu, J.-M.; et al. Deglycosylation of PD-L1 by 2-deoxyglucose reverses PARP inhibitor-induced immunosuppression in triple-negative breast cancer. Am. J. Cancer Res. 2018, 8, 1837–1846. [Google Scholar]
  79. Yélamos, J.; Moreno-Lama, L.; Jimeno, J.; Ali, S.O. Immunomodulatory Roles of PARP-1 and PARP-2: Impact on PARP-Centered Cancer Therapies. Cancers 2020, 12, 392. [Google Scholar] [CrossRef] [Green Version]
  80. Lampert, E.J.; Zimmer, A.; Padget, M.; Cimino-Mathews, A.; Nair, J.R.; Liu, Y.; Swisher, E.M.; Hodge, J.W.; Nixon, A.B.; Nichols, E.; et al. Combination of PARP Inhibitor Olaparib, and PD-L1 Inhibitor Durvalumab, in Recurrent Ovarian Cancer: A Proof-of-Concept Phase II Study. Clin. Cancer Res. 2020, 26, 4268–4279. [Google Scholar] [CrossRef]
  81. Karzai, F.; VanderWeele, D.; Madan, R.A.; Owens, H.; Cordes, L.M.; Hankin, A.; Couvillon, A.; Nichols, E.; Bilusic, M.; Beshiri, M.L.; et al. Activity of durvalumab plus olaparib in metastatic castration-resistant prostate cancer in men with and without DNA damage repair mutations. J. Immunother. Cancer 2018, 6, 141. [Google Scholar] [CrossRef] [PubMed]
  82. Domchek, S.M.; Postel-Vinay, S.; Im, S.-A.; Park, Y.H.; Delord, J.-P.; Italiano, A.; Alexandre, J.; You, B.; Bastian, S.; Krebs, M.G.; et al. Olaparib and durvalumab in patients with germline BRCA-mutated metastatic breast cancer (MEDIOLA): An open-label, multicentre, phase 1/2, basket study. Lancet Oncol. 2020, 21, 1155–1164. [Google Scholar] [CrossRef]
  83. Pusztai, L.; Yau, C.; Wolf, D.M.; Han, H.S.; Du, L.; Wallace, A.M.; String-Reasor, E.; Boughey, J.C.; Chien, A.J.; Elias, A.D.; et al. Durvalumab with olaparib and paclitaxel for high-risk HER2-negative stage II/III breast cancer: Results from the adaptively randomized I-SPY2 trial. Cancer Cell 2021, 39, 989–998.e5. [Google Scholar] [CrossRef]
  84. Vinayak, S.; Tolaney, S.M.; Schwartzberg, L.; Mita, M.; McCann, G.; Tan, A.R.; Wahner-Hendrickson, A.E.; Forero, A.; Anders, C.; Wulf, G.M.; et al. Open-label Clinical Trial of Niraparib Combined with Pembrolizumab for Treatment of Advanced or Metastatic Triple-Negative Breast Cancer. JAMA Oncol. 2019, 5, 1132–1140. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  85. Ramalingam, S.S.; Thara, E.; Awad, M.M.; Dowlati, A.; Haque, B.; Stinchcombe, T.E.; Dy, G.K.; Spigel, D.R.; Lu, S.; Lyer Singh, N. JASPER: Phase 2 trial of first-line niraparib plus pembrolizumab in patients with advanced non-small cell lung cancer. Cancer 2021. [Google Scholar] [CrossRef]
  86. Konstantinopoulos, P.A.; Waggoner, S.; Vidal, G.A.; Mita, M.; Moroney, J.W.; Holloway, R.; van Le, L.; Sachdev, J.C.; Chapman-Davis, E.; Colon-Otero, G.; et al. Single-Arm Phases 1 and 2 Trial of Niraparib in Combination with Pembrolizumab in Patients With Recurrent Platinum-Resistant Ovarian Carcinoma. JAMA Oncol. 2019, 5, 1141–1149. [Google Scholar] [CrossRef] [Green Version]
  87. Färkkilä, A.; Gulhan, D.C.; Casado, J.; Jacobson, C.A.; Nguyen, H.; Kochupurakkal, B.; Maliga, Z.; Yapp, C.; Chen, Y.A.; Schapiro, D.; et al. Immunogenomic profiling determines responses to combined PARP and PD-1 inhibition in ovarian cancer. Nat. Commun. 2020, 11, 1459. [Google Scholar] [CrossRef] [Green Version]
  88. Meng, X.W.; Koh, B.D.; Zhang, J.-S.; Flatten, K.S.; Schneider, P.A.; Billadeau, D.D.; Hess, A.D.; Smith, B.D.; Karp, J.E.; Kaufmann, S.H. Poly(ADP-ribose) Polymerase Inhibitors Sensitize Cancer Cells to Death Receptor-mediated Apoptosis by Enhancing Death Receptor Expression. J. Biol. Chem. 2014, 289, 20543–20558. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  89. Fenerty, K.E.; Padget, M.; Wolfson, B.; Gameiro, S.R.; Su, Z.; Lee, J.H.; Rabizadeh, S.; Soon-Shiong, P.; Hodge, J.W. Immunotherapy utilizing the combination of natural killer- and antibody dependent cellular cytotoxicity (ADCC)-mediating agents with poly (ADP-ribose) polymerase (PARP) inhibition. J. Immunother. Cancer 2018, 6, 133. [Google Scholar] [CrossRef] [PubMed]
  90. Guerra, N.; Tan, Y.X.; Joncker, N.T.; Choy, A.; Gallardo, F.; Xiong, N.; Knoblaugh, S.; Cado, D.; Greenberg, N.R.; Raulet, D.H. NKG2D-Deficient Mice Are Defective in Tumor Surveillance in Models of Spontaneous Malignancy. Immunity 2008, 28, 571–580. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  91. Raulet, D.H.; Guerra, N. Oncogenic stress sensed by the immune system: Role of natural killer cell receptors. Nat. Rev. Immunol. 2009, 9, 568–580. [Google Scholar] [CrossRef] [Green Version]
  92. Lam, A.R.; le Bert, N.; Ho, S.S.W.; Shen, Y.J.; Tang, M.L.F.; Xiong, G.M.; Croxford, J.L.; Koo, C.X.; Ishii, K.J.; Akira, S.; et al. RAE1 Ligands for the NKG2D Receptor Are Regulated by STING-Dependent DNA Sensor Pathways in Lymphoma. Cancer Res. 2014, 74, 2193–2203. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  93. Zhu, Z.-C.; Bai, Y.; Lu, X.-Z.; Qi, C.-J. Effects and Mechanism of PARP Inhibitor Olaparib on the Expression of NKG2D Ligands in HL-60 Cells. Zhongguo Shi Yan Xue Ye Xue Za Zhi 2020, 28, 1826–1830. [Google Scholar] [PubMed]
  94. Soriani, A.; Zingoni, A.; Cerboni, C.; Iannitto, M.L.; Ricciardi, M.R.; di Gialleonardo, V.; Cippitelli, M.; Fionda, C.; Petrucci, M.T.; Guarini, A.; et al. ATM-ATR–dependent up-regulation of DNAM-1 and NKG2D ligands on multiple myeloma cells by therapeutic agents results in enhanced NK-cell susceptibility and is associated with a senescent phenotype. Blood 2009, 113, 3503–3511. [Google Scholar] [CrossRef] [Green Version]
  95. Friedman, J.; Morisada, M.; Sun, L.; Moore, E.C.; Padget, M.; Hodge, J.W.; Schlom, J.; Gameiro, S.R.; Allen, C.T. Inhibition of WEE1 kinase and cell cycle checkpoint activation sensitizes head and neck cancers to natural killer cell therapies. J. Immunother. Cancer 2018, 6, 59. [Google Scholar] [CrossRef] [Green Version]
Figure 1. DDR-targeted agents that have been clinically approved or are in clinical trials. PARP-1 and PARP-2 become enzymatically active upon recognition of DNA breaks, cleave β-NAD+ and transfer ADP-ribose moieties (PARylation) onto specific amino acid residues of acceptor proteins. The kinases ATR and ATM phosphorylate hundreds of proteins, including CHK1 and CHK2, respectively, which are key mediators of the checkpoint function. Whereas ATM is activated by DNA double-strand breaks (DSBs), ATR is activated by DNA single-strand breaks (SSBs). The kinase DNA-PK is critical for DSB repair via the non-homologous end-joining pathway. The kinase WEE1 controls mitotic entry.
Figure 1. DDR-targeted agents that have been clinically approved or are in clinical trials. PARP-1 and PARP-2 become enzymatically active upon recognition of DNA breaks, cleave β-NAD+ and transfer ADP-ribose moieties (PARylation) onto specific amino acid residues of acceptor proteins. The kinases ATR and ATM phosphorylate hundreds of proteins, including CHK1 and CHK2, respectively, which are key mediators of the checkpoint function. Whereas ATM is activated by DNA double-strand breaks (DSBs), ATR is activated by DNA single-strand breaks (SSBs). The kinase DNA-PK is critical for DSB repair via the non-homologous end-joining pathway. The kinase WEE1 controls mitotic entry.
Cancers 13 06008 g001
Figure 2. Effects of DDR-targeted agents on the cell components of the immune system. DDR-targeted agents can affect different cells of the immune system, either with immunosuppressive functions (left) or effector functions (right), and thus contribute to promoting or inhibiting tumour progression. CSF1R, colony-stimulating factor 1 receptor; M1, M1 macrophages; M2, M2 macrophages; MDSC, myeloid-derived suppressor cells; NK, natural killer cell; PD-1, programmed death protein-1; PD-L1, programmed death ligand-1; Treg, regulatory T cells.
Figure 2. Effects of DDR-targeted agents on the cell components of the immune system. DDR-targeted agents can affect different cells of the immune system, either with immunosuppressive functions (left) or effector functions (right), and thus contribute to promoting or inhibiting tumour progression. CSF1R, colony-stimulating factor 1 receptor; M1, M1 macrophages; M2, M2 macrophages; MDSC, myeloid-derived suppressor cells; NK, natural killer cell; PD-1, programmed death protein-1; PD-L1, programmed death ligand-1; Treg, regulatory T cells.
Cancers 13 06008 g002
Figure 3. Schematic representation of the tumour microenvironment depicting the effects of DDR-targeted agents on the crosstalk between tumour cells and infiltrating immune cells. cGAS, GMP-AMP (cGAMP) synthase; IFN, interferon; NK, natural killer cell; NKG2DL, NKG2D ligand; PD-1, programmed death protein-1; PD-L1, programmed death ligand-1; pIRF3, phosphor IFN regulatory factor 3; STING, stimulator of IFN genes; Treg, regulatory T cells; TRAILR, TRAIL receptor.
Figure 3. Schematic representation of the tumour microenvironment depicting the effects of DDR-targeted agents on the crosstalk between tumour cells and infiltrating immune cells. cGAS, GMP-AMP (cGAMP) synthase; IFN, interferon; NK, natural killer cell; NKG2DL, NKG2D ligand; PD-1, programmed death protein-1; PD-L1, programmed death ligand-1; pIRF3, phosphor IFN regulatory factor 3; STING, stimulator of IFN genes; Treg, regulatory T cells; TRAILR, TRAIL receptor.
Cancers 13 06008 g003
Table 1. Phase II clinical trials combining agents targeting DNA damage response with immune checkpoint inhibitors (ClinicalTrials.gov; accessed on 1 October 2021).
Table 1. Phase II clinical trials combining agents targeting DNA damage response with immune checkpoint inhibitors (ClinicalTrials.gov; accessed on 1 October 2021).
Study
Identifier
Study DesignConditionTargetDrugPrimary Outcome
NCT03955471Single ArmOvarian NeoplasmsPARPOlaparib + TSR-042 (anti-PD-1 mAb)ORR (5 years)
NCT03951415Single ArmEndometrial Neoplasms, Uterine
Neoplasms
PARPOlaparib + Durvalumab (anti-PD-L1 mAb)PFS (12 weeks)
NCT03851614Two ArmsMismatch Repair Proficient Colorectal Cancer, Pancreatic Adenocarcinoma, LeiomyosarcomaPARPOlaparib + Durvalumab (anti-PD-L1 mAb)Changes in genomic and immune biomarkers (3 years)
NCT03780608Single Arm with 2 CohortsGastric Adenocarcinoma, Malignant MelanomaATRCeralasertib [AZD6738] + Durvalumab (anti-PD-L1
mAb)
ORR (2 years)
NCT03574779Single ArmOvarian NeoplasmsPARPNiraparib + TSR-042 (anti-PD-1 mAb) +
Bevacizumab (anti-VEGF mAb)
ORR (6 years)
NCT03565991Single Arm with 2 CohortsAdvanced Solid Tumours with Defects in BRCA1/BRCA2, Advanced Solid Tumours with Defects in ATMPARPTalazoparib + Avelumab (anti-PD-L1 mAb)OR (2 years)
NCT03330405Two Arms with 10 CohortsLocally Advanced or Metastatic Solid TumorsPARPTalazoparib + Avelumab (anti-PD-L1 mAb)ORR (2 years)
NCT03167619Two ArmsTriple Negative Breast CancerPARPOlaparib + Durvalumab (anti-PD-L1 mAb)PFS (12 months)
NCT02953457Single ArmRecurrent ovarian, fallopian tube, or primary peritoneal cancer with BRCA1 or BRCA2 genetic mutationPARPOlaparib + Durvalumab (anti-PD-L1 mAb) + Tremelimumab (anti-CTLA-4 mAb)PFS (3 and 6 months)
NCT02657889Single ArmTriple Negative Breast Cancer, Ovarian Cancer, Fallopian Tube Cancer, Peritoneal CancerPARPNiraparib + Pembrolizumab (anti-PD-1 mAb)ORR (40 weeks)
NCT02571725Single ArmOvarian Cancer, Fallopian Tube Cancer, Peritoneal NeoplasmsPARPOlaparib + Tremelimumab (anti-CTLA-4 mAb)ORR (2 years)
mAb: monoclonal antibody; ORR: overall response rate; PFS: progression-free survival; OR: objective response.
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Share and Cite

MDPI and ACS Style

Lutfi, N.; Galindo-Campos, M.A.; Yélamos, J. Impact of DNA Damage Response—Targeted Therapies on the Immune Response to Tumours. Cancers 2021, 13, 6008. https://doi.org/10.3390/cancers13236008

AMA Style

Lutfi N, Galindo-Campos MA, Yélamos J. Impact of DNA Damage Response—Targeted Therapies on the Immune Response to Tumours. Cancers. 2021; 13(23):6008. https://doi.org/10.3390/cancers13236008

Chicago/Turabian Style

Lutfi, Nura, Miguel Alejandro Galindo-Campos, and José Yélamos. 2021. "Impact of DNA Damage Response—Targeted Therapies on the Immune Response to Tumours" Cancers 13, no. 23: 6008. https://doi.org/10.3390/cancers13236008

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