Next Article in Journal
Computation-Aided Design of Albumin Affibody-Inserted Antibody Fragment for the Prolonged Serum Half-Life
Previous Article in Journal
Non-Invasive Physical Plasma for Preventing Radiation Dermatitis in Breast Cancer: A First-In-Human Feasibility Study
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Immune Modulatory Effects of Molecularly Targeted Therapy and Its Repurposed Usage in Cancer Immunotherapy

1
Department of Liver Surgery and Transplantation, Key Laboratory of Carcinogenesis and Cancer Invasion (Ministry of Education), Liver Cancer Institute, Zhongshan Hospital, Fudan University, Shanghai 200032, China
2
Key Laboratory of Medical Epigenetics and Metabolism, Institutes of Biomedical Sciences, Fudan University, Shanghai 200032, China
3
State Key Laboratory of Genetic Engineering, Fudan University, Shanghai 200433, China
*
Author to whom correspondence should be addressed.
These authors contributed equally to this work.
Pharmaceutics 2022, 14(9), 1768; https://doi.org/10.3390/pharmaceutics14091768
Submission received: 9 July 2022 / Revised: 13 August 2022 / Accepted: 22 August 2022 / Published: 24 August 2022

Abstract

:
The fast evolution of anti-tumor agents embodies a deeper understanding of cancer pathogenesis. To date, chemotherapy, targeted therapy, and immunotherapy are three pillars of the paradigm for cancer treatment. The success of immune checkpoint inhibitors (ICIs) implies that reinstatement of immunity can efficiently control tumor growth, invasion, and metastasis. However, only a fraction of patients benefit from ICI therapy, which turns the spotlight on developing safe therapeutic strategies to overcome the problem of an unsatisfactory response. Molecular-targeted agents were designed to eliminate cancer cells with oncogenic mutations or transcriptional targets. Intriguingly, accumulating shreds of evidence demonstrate the immunostimulatory or immunosuppressive capacity of targeted agents. By virtue of the high attrition rate and cost of new immunotherapy exploration, drug repurposing may be a promising approach to discovering combination strategies to improve response to immunotherapy. Indeed, many clinical trials investigating the safety and efficacy of the combination of targeted agents and immunotherapy have been completed. Here, we review and discuss the effects of targeted anticancer agents on the tumor immune microenvironment and explore their potential repurposed usage in cancer immunotherapy.

1. Introduction

Cancer has been a major leading cause of death worldwide [1]. The novel technology allowed researchers to focus on cancer initiation and progression at levels of cellular and molecular phenotype. Furthermore, the concept of hallmarks of cancer attracted attention to the commonalities that are shared among distinct types of cancer, and many drugs were exploited based on these characteristics. Cancer pharmacological treatments can range from chemotherapy to targeted therapy and immunotherapy. In the 1940s, chemotherapeutic drugs raised hope for patients with advanced or metastatic cancer. However, chemotherapy’s toxicity to normal cells plagued clinical doctors for decades until the approval of targeted anticancer agents [2]. Molecularly targeted therapy is a type of cancer treatment to inhibit cancer progression via small-molecule drugs or antibodies [3]. For instance, the anti-angiogenesis agents have been exploited and approved for treating solid tumors, a milestone in the discovery history of molecularly targeted agents [4].
Targeted agents have become first- or second-line treatments for most advanced malignancies, including breast cancer, lung cancer, colorectal carcinoma (CRC), hepatocellular carcinoma (HCC), renal cell carcinoma (RCC), and others [5]. Targeted agents exert broad anti-tumor activity via direct inhibition on tumor cells and indirect impacts on the tumor microenvironment. Agents approved in clinical use mainly act by inhibiting cycle-dependent kinase (CDK), KRAS, and PI3K signaling, DNA damage repair (DDR) and apoptosis, and ErbB family signaling [6]. In addition, many drugs do not belong to the above four categories but are widely used in clinical treatment, such as the Bruton tyrosine kinase (BTK) inhibitor. During cancer cell invasion and spread, multiple signaling pathways are involved. Indeed, co-targeting different molecules induced synergetic effects, and multi-targeted drugs showed a significant clinical advantage [7]. However, drug resistance caused by targeted agents results in limited response rates and duration of response, especially for patients with advanced or aggressive malignancies. For instance, MET amplification has been proven to be the mechanism of primary resistance to epidermal growth factor receptor (EGFR) tyrosine kinase inhibitor (TKI) therapy in EGFR-mutant non-small cell lung cancer (NSCLC) patients.
Beyond blocking oncogenic mutations or transcriptional targets, molecularly targeted agents modulate the immune context in the tumor microenvironment, also known as the tumor immune microenvironment (TIME). For example, CDK4/6 inhibitors “inflame” TIME by recruiting immune effector cells and suppressing Treg cell proliferation. With a deeper understanding of various subsets of immune cells, there has been a recent surge of interest in exploring the role of TIME in tumorigenesis and metastasis. As an example, to evade the attack of effector immune cells, tumor cells can produce immune suppressive factors, such as IL-6, IL-8, and transforming growth factor-β (TGF-β) to recruit immunosuppressive cells and impair the anti-tumor immune response, favoring tumor progression [8,9]. Thus, immunotherapy is based on the studies of how immunity recognizes and eliminates cancer cells and the mechanism of cancer cells’ evolution to avoid the attack. The identification of immune checkpoints in TIME, to some extent, reveals the mechanism that tumor-specific T cells cannot eliminate cancer cells efficiently without pharmaceutical interventions.
The clinical success of ICIs has brought a revolution in cancer therapy, and it is widely used as a standard treatment in various cancers. Until now, the targets of FDA-approved agents focus on CTLA-4 (ipilimumab), PD-1 (pembrolizumab, nivolumab, cemiplimab, etc.), PD-L1 (atezolizumab, durvalumab, avelumab), and LAG-3 (relatlimab) [10,11]. Despite the great progress in recent years, using ICIs as monotherapy is still limited due to an unsatisfactory response rate. Based on preclinical and clinical data, it is interesting that most targeted anticancer agents could enhance the patient response to ICIs. For example, venetoclax, the first FDA-approved BCL2 inhibitor, increased the T effector memory cells and showed great potential in combination with ICIs [12]. What is more, according to the results of IMbrave150, a phase 3 clinical trial, the FDA approved the combination of atezolizumab, selectively targeting PD-L1, and bevacizumab, a VEGF-A-targeting monoclonal antibody, for first-line treatment in patients with unresectable or metastatic HCC [13]. In contrast, KEYNOTE-240, another phase 3 clinical trial, tested the efficacy and safety of pembrolizumab, an anti-PD-1 monoclonal antibody, as monotherapy for advanced HCC, and its result did not reach statistical significance [14].
The existing molecularly targeted agents are designed to block the signaling pathways involved in hallmarks of cancer, such as inducing angiogenesis, immune evasion, and metabolic reprogramming. It is not surprising that the repurposing of targeted therapy combined with immunotherapy may become the future paradigm and direction of clinical investigation. In this review, we summarize how the targeted anticancer agents influence the tumor microenvironment and the potential combination of targeted therapy with immunotherapy.

2. Modulatory Effects of Targeted Therapy on Immune Cells

TIME plays a vital role in the modulation of tumor initiation, progression, and drug resistance. The generation of immune response to cancer is a multistep process and the last step is to eliminate cancer cells, which occurs in TIME and is an important rate-limiting step. Using ICIs released the restriction of effector T cells, and thereby many cancer patients benefit from this approach [10,15]. However, a few cancer types, such as pancreatic ductal adenocarcinoma (PDAC), neuroendocrine neoplasm, and mismatch repair-proficient CRC, barely benefit from immunotherapy, which may be due to the “cold” immune phenotype that contains fewer effector immune cells and more immunosuppressive cells [10,16]. It is possible to reuse targeted drugs to inhibit the specific type of cells, molecules, or pathways in TIME, enhancing the efficacy of immunotherapy. An in-depth investigation of targeted anticancer agents’ immunosuppressive and immunostimulatory capacity may provide the theoretical basis for the combinatorial treatment with immunotherapy (Figure 1).

2.1. Effector T Cells

Cytotoxic CD8+ T cell is the key to adaptive immunity against tumor cells. Notably, eradicating tumor cells by effector T cells is a multistep process; many immunosuppressive factors directly or indirectly counteract this process. For example, activated T cells in TIME obtain energy through glycolysis even in the presence of oxygen after metabolic reprogramming, and the block of T cells glycolysis attenuates their anti-tumor activities [17]. A subset of T cells in a state of exhaustion is defined as the loss of effector function, such as cytotoxicity, and correlates with the resistance to immunotherapy. Consistent with these notions, improving the infiltration of tumor-specific T cells, maintaining activated T cells metabolism, reversing effector T cells exhaustion, and activating T cells’ tumor-killing capacity have long been the purposes of developing new therapeutic strategies.
To date, three CDK4/6 inhibitors, including palbociclib, ribociclib, and abemaciclib, have been approved by FDA. Clinical trials demonstrate that CDK4/6 inhibitors prolong the progression-free survival in patients with estrogen receptor (ER)-positive breast cancer when combined with anti-estrogen therapy. The immunostimulatory activity of CDK4/6 inhibition can be achieved by interacting with T cells via the depression of the nuclear factor of the activated T cell (NFATC) family [18]. In the TIME of tumors treated with the BRAF inhibitor, anti-tumor-specific CD8+ T cells were enriched due to increased tumor antigen expression [19,20]. Beyond using BRAF inhibitors singly, the BRAF(V600E/K)-mutant melanoma patients benefited from the combinational treatments of BRAF inhibitor and MEK inhibitor [21]. It has been proved that the immunoregulatory effects induced by combinatorial therapy are vital to tumor shrinkage [22]. This notion is supported by the combination of BRAF and MEK inhibition, which boosts the expansion of tumor-specific T cells and the abundance of TCR repertoire, which is due to the induction of TCF7 and T-bet [23,24]. Pharmacological inhibition of MDM2 by ALRN-6924 improves the infiltration of cytotoxic CD8+ T cells, reflecting the potential for MDM2 inhibitors combined with immunotherapy [25]. The existing evidence suggests that VEGF/VEGFR signaling decreases the proliferation of T cells and increases the expression of PD-1 on CD8+ T cells [26]. In both lung and CRC murine models, BD0801, an anti-VEGF mAb, showed a significant anti-tumor capacity when combined with ICIs, whose potential mechanism is increasing CD8+ effector T cells and reducing CD8+PD-1+ exhaustion T cells [27]. Sorafenib and Lenvatinib, two multi-kinase inhibitors, are the first-line drugs for advanced HCC [28]. In the mouse HCC model, sorafenib showed the ability to deplete PD-1+CD8+ T cells, enhancing the anti-tumor immune response [29]. Indeed, the clinical data showed that the number of CD8+Ki67+IFN-γ+ T cells increased after treatment with sorafenib, and the upregulation of CD8+Ki67+IFN-γ+ T cells was associated with better patient outcomes [30]. Lenvatinib has been proven to augment anti-tumor activities by increasing T cell infiltration and decreasing the number and immunosuppression of myeloid-derived suppressor cells [31]. Cancers harboring epidermal growth factor receptor (EGFR) mutation always build a non-inflamed TIME [32]. The treatment with EGFR inhibitor remodels the TIME by improving the numbers of T cells and decreasing the immunosuppressive cells [33]. Conversely, in the breast cancer mouse model, the combination of PARP1 and PARP2 resulted in remolding the TIME and less activation of T cells [34]. Altogether, these findings suggest that targeted drugs could modulate the effector T cells priming, differentiation, and functions. Notably, among quantities of targeted molecules, the agents targeting ErbB augment T cell-based anti-tumor activities from multiple aspects, such as increasing the number of CD8+ T cells, enhancing cytotoxic T cell function, and reducing T cell exhaustion, especially in NSCLCs and CRCs [35].

2.2. Regulatory T Cells

Regulatory T cells (Treg) control autoimmunity and suppress the inflammation response, which maintains the hosts’ immune homeostasis. However, in TIME, Treg cells cooperate with other immunosuppressive cells to promote tumor growth and metastasis. It has been proven that chemotherapy such as cyclophosphamide (CTX) can obliterate Treg cells and augment anti-tumor activities [36]. The low dose of CTX attenuated the suppression of immunity induced by CD4+CD25+ Treg cells. It promoted the aggregation of activated T cells, which has been used as the pretreatment of a cancer vaccine [37,38]. The mechanism of immune invasion that progressive cancer recruits Treg cells could be understood as a host’s “self-identity” to tumor cells. A deeper insight into the biology of Treg cells reveals that using agents targeting the critical process of Treg cell function is a promising strategy.
The effect of CDK4/6 inhibitors on Treg cells has been tested both in vitro and in vivo. The underlying mechanism for the depletion of Treg cells is the overexpression of p21 and repression of DNA methyltransferase 1 (DNMT1) [39]. From the analysis of RIBECCA trial, ribociclib, a CDK4/6 inhibitor, significantly downregulated peripheral CD4+FOXP3+CD25+ Treg cells [40]. Beyond increasing T cell infiltration, another potential mechanism of lenvatinib-mediated immunoenhancement is reducing tumor intrinsic Treg cells, which opens up an opportunity to combine it with immunotherapy [41]. Moreover, the selective-PI3K inhibitor ZSTK474, in an optimal dose, deletes Treg cells specifically, and, intriguingly, PI3K inhibition promoted the differentiation into tumor-specific CD8+ memory T cells, which contributed to a more robust immune response [42]. A recent clinical study showed that intermittent use of AMG319, a novel PI3Kδ inhibitor, may result in anti-tumor immunity by reducing intratumoral Treg cells with controllable adverse events [43]. The evidence showed that aberrant EGFR signaling increased the frequency of Treg cells in TIME by chemokine production, therefore EGFR inhibitors suppressed this process, improving the response of ICIs in NSCLC patients [35]. Altogether, the evidence that Treg cells support tumor progression and restraining Treg cells or their function leads to tumor regress argues that these molecules or pathways could be promising targets. However, it is important to note that the high frequency of Treg cells correlated with a good prognosis in ER- breast cancer and mismatch repair-proficient CRC [44,45,46].

2.3. B Cells

Depending on the composition of the tumor microenvironment, tumor-infiltrating B cells (TIBs) play the opposite role of suppressing or promoting cancer cells. TIBs can exert anti-tumor ability by producing the tumor-specific antibody, assisting T cell priming and activation, and killing cancer cells directly [47]. B cells are critical to the formation of the tumor-associated tertiary lymphoid structure (TLS), which supports the anti-tumor activity and may suggest better patient outcomes [48]. IL-10 is considered an immunosuppression cytokine, and the subset of B cells that produce IL-10 is recognized as regulatory B cells (Breg). Unlike Foxp3, the marker of Treg cells, the definitive marker of Breg cells is still lacking. Bregs can induce an immune-suppressive milieu in TIME by inhibiting cytotoxic T cell or cytokine secretion.
Ibrutinib is a covalent and irreversible BTK inhibitor and has been approved to treat mantel cell lymphoma and chronic lymphatic leukemia [49]. Ibrutinib breaks the cross-talk between B cells and FcRγ+ macrophages, activating T cell-based antitumor response in PDAC mouse models [50]. Targeting CDK4/6 enhanced antitumor immunity by inducing a pro-inflammatory environment, where the recruitment of B cells was impressive [51]. This is attributed to CXCL13 secreted by abemaciclib-treated mouse ovarian cancer cells, which support B cell homing to follicles and subsequent T cell activation [51]. Studies on the effects of targeted anticancer agents on TIBs are insufficient. Existing evidence emphasizes the importance of TLS in response to immunotherapy [52,53]. Correspondingly, understanding how anticancer agents influence B cell infiltration and formation of TLS is crucial for further studies.

2.4. Natural Killer (NK) Cells

Natural killer (NK) cells play an essential role in innate immunity, which acts as the “killer” to control tumor growth and metastasis. Based on their surface expression level of CD56, NK cells are usually divided into two subsets, including CD56dim NK cells and CD56bright NK cells [54,55]. NK cells participate in innate and adaptive immune responses, and their anti-tumor activities do not require antigen-sensitization and are not limited by the major histocompatibility complex (MHC). In addition, the chimeric antigen receptor (CAR) technology used in NK cells shows more advantages than in T cells, such as a broader range of sources of cells and a lower incidence of therapy toxicity [56]. Targeting NK cells could therefore generate a new promising approach to augmenting anti-tumor treatment.
In murine models of KRAS-mutant lung cancer, combined MEK and CDK4/6 inhibition trigger innate immune response, especially by NK cells [57]. Interestingly, retinoblastoma (RB)-mediated cellular senescence contributed to NK cell-mediated cytotoxicity, and this study indicates that specific targeted anticancer agents could remodel the disabled immune surveillance [57]. While there are no selective inhibitors to CDK8, NK cell-specific CDK8 deletion promotes the secretion of perforin in vitro. It improves the outcomes in models of melanoma, lymphoma, and leukemia [58]. Tumor necrosis factor (TNF)-related apoptosis-inducing ligand (TRAIL) could upregulate the percentage of NK cells in TIME when used in combination with CDK inhibitor in NSCLC mouse models [59]. Supporting the efficacy of the BRAF inhibitor against BRAF(V600E)-mutant melanoma, a recent study suggested that BRAF inhibition directly enhanced NK cells’ immunostimulatory capacity via upregulating CD69 expression and IFNR release [60]. Ovarian cancer cells pre-treated by EGFR TKIs are more sensitive to NK cell-mediated antibody-dependent cell-mediated cytotoxicity (ADCC), which points to an exciting possibility of designing new combined immunotherapy [61]. Conversely, the expression of an NK-cell-activating ligand, MICA, and DNAM-1, was downregulated after the treatment of vemurafenib [62]. PI3K∂-selective inhibitor idelalisib attenuated the proliferation and cytotoxicity of NK and T cells, potentially explaining the limited efficacy of idelalisib [63]. The histone deacetylase inhibitor (HDACi) has been used to treat several hematological malignancies. However, HDACi upregulated NKG2DLs on tumor cells in vitro and impaired NK cells’ viability in vivo [64]. Further works about targeted drugs’ exact effects on NK cells can accelerate the rational development of NK cell-based immunotherapy.

2.5. Neutrophils

Neutrophils, as fast-response immune cells, obliterate different kinds of pathogens but may play both pro-and anti-tumor roles in the TIME. By virtue of different functions, neutrophils are classified using several terms, including N1/N2 neutrophils, tumor-associated neutrophils (TANs), and polymorphonuclear neutrophil myeloid-derived suppressor cells (PMN-MDSCs) [65].
Because of the side effects in normal organs, the use of TGF-β inhibitors is limited. Interestingly, neutrophils are utilized as vehicles to load TGF-β inhibitors and then transport drugs to tumor sites [66]. Nanomedicine that can locally release TGF-β was developed and showed potential in boosting neutrophils from the N2 to N1 phenotype in vitro and in vivo [67]. Conversely, a high dose of VEGFR2 inhibitor, apatinib, induced the expression of IL17A by γδ T cells, which resulted in neutrophil polarization to N2 phenotype and the exhaustion of CD8+ T cells in breast cancer mouse models. VEGFR inhibitors boosted the migration of neutrophils, and neutrophils could establish the pre-metastasis niche in favor of circulating tumor cells to seed [68].
Recent studies highlight that driving reverse migration is a more reasonable strategy than the depletion of neutrophils and preventing neutrophil infiltration [69,70]. Regarding the plasticity of neutrophils in TIME, finding specific drugs that induce the transformation of N2 to N1 TANs is expected to be another direction. Single-cell analysis of PDAC patients’ samples demonstrates that pro-tumor TANs are prone to exhibiting high glycolytic activity. Cancer cells can induce the glycolytic switch of TANs to mediate immunosuppressive TIME, which may open up a new mode of therapy. Altogether, with a much clearer understanding of TAN biology, we may anticipate benefits from molecularly targeted drugs that modulate TANs.

2.6. Dendritic Cells

Dendritic cells (DCs) function in antigen recognition and presentation and initiate an adaptive immune response [68]. While cancer cells present antigens onto their major histocompatibility complex I (MHCI), it is vital that DCs cross-present the tumor antigens to prime CD8+ T cells. Taking into account the latest therapy strategies, DC vaccines show potent tumor growth inhibition [71].
Trastuzumab, targeted to the extracellular domain of HER2, mediated immunostimulatory based on recognition and cross-presentation of tumor antigen by DCs, which was especially realized through Fc receptor-mediated mechanism [72]. Exiting evidence implies that various PI3K isoforms regulate DCs’ functions [73]. A pan-PI3K inhibitor, copanisib, dramatically upregulated CD80, CD86, MHC-I, and MHC-II in DCs, and its combination with ICIs showed a potent antitumor response in bladder cancer mouse models [74]. A combination of BRAF and MEK inhibition promoted the maturation of DCs and T cell activation, mainly reflecting the potential of their combination with immunotherapy [24]. Currently, plenty of DC-based immunotherapies have been developed and tested in clinical trials. It is a great supplement to discovering targeted agents that modulate DCs’ anti-tumor capacity to improve DC maturation and antigen presentation.

2.7. Macrophages

Traditionally, macrophages are roughly divided into antitumor (M1) or protumor (M2) categories. With the development of single-cell technology, it has been realized that dynamic changes in macrophage phenotypes occur during tumorigenesis and progression, and different TAMs subpopulations are responsible for distinct functions [75]. The molecularly targeted agents that inhibit or alert TAMs pro-tumoral activities need extensive investigation.
Blocking the recruitment of macrophages is an efficient approach to deplete TAMs. VEGF contributes to the attraction of monocytes by VEGFR-1 and promotes angiogenesis. Surprisingly, anti-VEGF therapy induced macrophages to move to the tumor via the establishment of tumor hypoxia [76]. Several studies have shown solid evidence that repolarizing macrophage leads to a promising direction to augment immunotherapy. In the murine CRC model, CD11b+ Ly6C+ MHC-II+ cells, an intermediate state from monocytes to TAMs, accumulated in the tumor environment, where MEK inhibition suppressed macrophage polarization [77]. Interestingly, CD11b+ FcγRI/III+ myeloid cells were discovered with increasing PI3Kγ signaling in PDAC [50], where PI3K inhibitor reprogramed macrophages and improved the response to chemotherapy [78]. Given the crucial role of P53 activation in macrophages, the MDM2 inhibitor reprogramed M1-like macrophages into M2-like macrophages in syngeneic mouse models of CRC and HCC [79].
Conversely, using EGFR targeting monoclonal antibodies may activate M2 macrophages, which promote tumor progression [80]. In the TIME of BRCA-associated triple-negative breast cancer (TNBC), a macrophage is a major population of immune cells. Multi-omics data showed that PARP inhibitors alter the anti-tumor activities of macrophages through metabolic reprogramming, which shapes an immunosuppressive environment, but provides a potential therapeutic strategy that targets TAMs in BRCA-associated TNBC [81]. In the future, investigating the dynamic polarization of TAMs and their functional phenotypes could significantly accelerate the discovery of novel therapeutic strategies.

2.8. Myeloid-Derived Suppressor Cells (MDSCs)

MDSCs exert all their skills to promote tumor growth and metastasis, such as dampening immune response and facilitating tumor angiogenesis, raising great interest in developing a therapeutic strategy that targets MDSCs. Studies have elucidated that MDSCs are observed in lymphoid tissues, bone marrow, and peripheral blood, in addition to TIME. MDSCs are classified into three categories: Monocytic-MDSCs (M-MDSCs), granulocytic-MDSCs (G-MDSCs), and early immature-MDSCs (eMDSCs) [82]. There is another classification based on morphological and phenotypical traits, and MDSCs are recognized as polymorphonuclear MDSC (PMH-MDSC) and monocytic MDSC (M-MDSC) [82].
Combined CDK 4/6 and PI3K inhibitors significantly depleted MDSCs in TNBC [83]. In a KRAS-driven murine lung cancer model, trametinib, a MEK inhibitor, directly depleted MDSCs [84]. The potential mechanism includes trametinib acting on the MEK pathway, which is necessary for the expansion of MDSCs, and trametinib’s influences on the secretion of osteopontin leading to MDSC depletion [84]. Combined BRAF, MEK, and CDK 4/6 inhibition showed promising anti-tumor activity, driven by reducing the population of tumor-intrinsic myeloid cells in syngeneic mouse models of melanomas [85]. A recent study indicated that the portion of G-MDSCs in peripheral blood was decreased with the treatment of bevacizumab-containing regimens compared with non-bevacizumab-based regimens in patients with NSCLC [86]. Targeted anticancer agents not only affect the number of MDSCs but also their immunosuppressive capacity. Treating mice with a VEGFR inhibitor, axitinib, reprogramed MDSCs toward an antigen-presenting phenotype [87]. Indeed, the approach to regulating myeloid cell differentiation to eliminate the immunosuppression phenotype is worth exploring.
Despite the immunostimulatory results, targeted drugs interfered with the anticancer response of immune cells. This may be one of the mechanisms that result in positive or negative outcomes in clinical research on combination treatment. However, both the immunostimulatory and immunosuppressive effects of targeted agents are not well understood. Further studies on how molecularly targeted therapy influences TIME are needed.

3. Modulatory Effects of Targeted Therapy on Immune Checkpoints

Different drugs have diverse impacts on the expression of immune checkpoints such as PD-1/PD-L1. A CDK4/6 inhibitor, Palbociclib, remodeled the TIME not only by its inhibition of RB phosphorylation but also by upregulating the PD-L1 protein level on tumor cells [88]. Consistent with this result, combined Palbociclib with ICIs regressed tumor growth and prolonged survival in mouse models of prostate cancer [88]. In addition, the PARP inhibitor inactivated GSK3β, which attenuated the proteasome degradation of PD-L1 in vitro and in vivo [89,90]. PARP inhibitors, Olaparib and Rucaparib, have been proven to upregulate PD-L1 expression in small and non-small cell lung cancer [91,92]. Another study also indicated that a DNA double-strand break due to treatment with PARPi was involved in the increased PD-L1 expression [93]. The evidence suggested that dabrafenib and trametinib, alone or in combination, were promising treatments for metastatic melanoma [94]. The upregulation of PD-L1 on melanoma cells was due to sustained exposure to dabrafenib, trametinib, or dabrafenib plus trametinib, and the increased level of PD-L1 may be a biomarker of acquired resistance to these drugs [95]. Conversely, cetuximab and erlotinib, EGFR inhibitors, blocked the tyrosine phosphorylation and suppressed the MAPK signaling, which led to the degradation of PD-L1 mRNA [96]. MEK1/2 inhibitors prevented IFNγ-induced PD-L1 mRNA upregulation, whose mechanism differs from the EGF-induced PD-L1degradation [96]. PRM2 promoted the expression of PD-L1 via the RRM2/ANXA1/AKT axis [97]. Though there is no evidence that the existing PRM2 inhibitors function effectively in anti-tumor activities [98,99], it offers a direction to develop a new PRM2 inhibitor. c-MET promoted the progression of HCC and the high expression of MET correlated with short patient survival [100,101]. However, a phase 3 study of the nonselective MET inhibitor tivantinib in patients with MET-high and previously treated advanced HCC showed that tivantinib did not improve overall survival [102]. A recent study demonstrates that MET showed a negative correlation with PD-L1 in HCC and prolonged the survival in HCC mouse models by simultaneously blocking MET and the PD-1/PD-L1 pathway [103].
Treatment with the MEK inhibitor in a pulsatile way showed better control of tumor proliferation than continuous treatment [104]. CTLA-4 expression was upregulated after the abovementioned treatment in vitro and in vivo [104]. Decitabine, a DNMT inhibitor, induced the up-regulation of CTLA-4 in a dose-dependent way [105]. Correspondingly, Decitabine combined with the CTLA-4 blockade enhanced the innate and adaptive immune response in murine ovarian cancer models [106]. Taken together, immune checkpoints are regulated by various molecularly targeted drugs in different cancer types. Thus, molecularly targeted agents’ cancer-specific immunomodulatory role needs further investigation, especially combined with immunotherapy.

4. Potential Combinations of Targeted Therapy and Immunotherapy

In the past decade, significant progress in developing targeted drugs and ICIs has enhanced the diversity of treatment regimens. Recent clinical trials have demonstrated that long-term tumor remission with tolerable side effects can be achieved by combining targeted therapy and immunotherapy.

4.1. CDK Inhibitors with ICIs

CDKs are a family of protein kinases involved in cell cycles, gene transcription, insulin secretion, glycogen synthesis, and neuronal functions, including 21 CDKs and 5 CDK-like genes [107]. The overexpression or deregulation of CDKs is generally considered to lead to unlimited proliferation and dysregulated bioactivity in several tumor types, such as breast cancer, CRC, and sarcoma [108,109]. Several CDK inhibitors were developed, among which the CDK4/6 inhibitors palbociclib, ribociclib, and abemaciclib were recommended as vital components of systemic therapy for HR+ breast cancer. In a recent phase I/II clinical trial (NCT02778685), combination therapy of palbociclib, pembrolizumab, and Letrozole showed good tolerance and a good objective response rate (ORR = 56%) as the first-line treatment for HR-positive metastatic breast cancer [110]. A phase Ib clinical trial (NCT02779751) evaluated the combination of abemaciclib and pembrolizumab in patients with NSCLC and HR+ and HER2− breast cancer. Results showed that abemaciclib plus pembrolizumab achieved a satisfactory ORR of 14.3% with a manageable safety profile in HR+ and HER2− breast cancer patients [111]. At the same time, the combination led to greater toxicity than each agent alone in NSCLC patients [112]. In summary, the contrary results in breast cancer and NSCLC patients demonstrated both anti-tumor activity and the possible toxicity of combination treatment, which requires more risk–benefit consideration (Table 1). To conclude, the combination of ICIs and CDK inhibitors may soon be applied in clinical use for breast cancer. More evidence is needed for lung cancer.

4.2. KRAS Signaling Inhibitors with ICIs

RAS is one of the most frequent mutations in human cancer and KRAS is the most frequently mutated isoforms, constituting approximately 86% of all RAS mutations [113]. Oncogenic KRAS mutation activates a series of downstream proteins, including PI3K, AKT, RAF, mTOR, and MAPK. Despite the strong oncogenic effect of KRAS in human cancer, KRAS mutants, except G12C, remain undruggable. Therefore, agents that target downstream signaling pathways are considered possible alternatives.
Mutated BRAF inhibitors dabrafenib and vemurafenib were approved for treating BRAF(V600E) metastatic melanoma [114,115]. Dabrafenib also showed clinical activity in NSCLC patients with BRAF(V600E) mutation [116]. Trametinib, a MEK1/2 inhibitor, showed antitumor activity against BRAF(V600E)-positive cancer [117]. Dabrafenib plus trametinib is now applied as one of the first-line therapies for advanced NSCLC or melanoma with a BRAF(V600E) mutation [118,119]. Intriguingly, a recent report illustrated that the activation of MAPK signaling is a compensatory mechanism of PI3K-Akt inhibition, leading to drug resistance to dabrafenib plus trametinib, and the co-administration of two pathways’ inhibitors shows synergistic anti-tumor activity [120,121]. A recent phase III clinical trial (NCT02224781) reported that the treatment of nivolumab plus ipilimumab followed by dabrafenib plus trametinib resulted in a higher 2-year OS (Overall Survival) rate (72% vs. 52%, respectively; p = 0.0095) than the treatment of dabrafenib plus trametinib followed by nivolumab and ipilimumab for patients with advanced BRAF(V600)-mutant melanoma [122]. A phase II clinical trial (NCT02130466) in BRAF-mutant melanoma showed that the triplet combination of dabrafenib, trametinib, and pembrolizumab had a longer median PFS (Progression-free Survival) (16.0 vs. 12.5 months, respectively; p = 0.043) and a higher response rate (59.8% vs. 27.8%, respectively) with a higher rate of grade 3/4 adverse events (58.3% vs. 26.7%, respectively) than the doublet combination of dabrafenib and trametinib [123]. A longer median PFS (16.2 vs. 12.0 months, respectively; p = 0.042) with a higher rate of grade 3 adverse events (55% vs. 33%) were also reported in dabrafenib, and trametinib plus spartalizumab than dabrafenib and trametinib for BRAF(V600)-mutant advance melanoma [123]. These results emphasized that adverse events could prevent the wide application of KRAS signaling inhibitors with ICIs, but the combination might be an alternative therapy under certain circumstances.
Cobimetinib, another MEK inhibitor, was approved for the treatment of metastatic melanoma in combination with vemurafenib based on a phase III clinical trial [124]. However, cobimetinib plus atezolizumab did not demonstrate a longer PFS than pembrolizumab alone in BRAF wild-type melanoma (5.5 vs. 5.7 months, respectively; p = 0.30), which showed that the combination of a MEK inhibitor and ICIs might not be more effective in these patients [125]. The phase III IMspire150 clinical trial (NCT02908672) investigated the possibility of the combination of cobimetinib, vemurafenib, and atezolizumab for advanced melanoma, and the median PFS was significantly prolonged in the triplet group than the conventional combination of cobimetinib and vemurafenib (15.1 vs. 10.6 months, respectively; p = 0.025) [126]. In biliary tract cancers, cobimetinib plus atezolizumab improved the median PFS (3.65 vs. 1.87 months, respectively; p = 0.027) compared with atezolizumab monotherapy based on a phase II trial (NCT03201458) [127]. A phase II study of cobimetinib plus atezolizumab for NSCLC patients is still ongoing (NCT03600701) [128]. Several other KRAS signaling inhibitors were approved for clinical use, including a PI3K inhibitor, alpelisib, and an mTOR inhibitor, everolimus. However, the safety and efficacy of these targeted agents in combination with ICIs remain unclear.

4.3. ErbB Family Inhibitors with ICIs

Overactivation of the ErbB protein family drives the tumorigenesis and development of various malignancies, including breast cancer, CRC, head and neck squamous cell carcinoma (HNSCC), and NSCLC [129]. The ErbB protein family consists of four receptor tyrosine kinase members: ErbB1/epidermal growth factor receptor (EGFR), ErbB2/HER2, ErbB3, and ErbB4. Over the past two decades, TKIs and monoclonal antibodies targeting EGFR and HER2 were developed and applied for the front- and subsequent-line treatments of human cancers.
Approved EGFR inhibitors include gefitinib, erlotinib, osimertinib, lapatinib, vandetanib, cetuximab, panitumumab, and necitumumab [130]. A phase I study (NCT02088112) reported that median PFS was 10.1 months, and ORR was 63.3% in NSCLC patients receiving gefitinib plus durvalumab [131]. No significant increase in PFS with higher toxicity was reported in the combination of gefitinib plus durvalumab. A phase I study (NCT02040064) reported that gefitinib plus tremelimumab limitedly improved survival (median PFS = 2.2 months) with a high adverse event rate [132]. In a phase I/II KEYNOTE-021 clinical trial (NCT02039674), gefitinib plus pembrolizumab was not feasible in NSCLC patients because 71.4% of patients had grade 3/4 liver toxicity [133]. Moreover, erlotinib plus pembrolizumab did not improve drug response compared with monotherapies. These results showed that a combination of gefitinib and ICIs might not be practicable due to side effects and that the combination of erlotinib and ICIs needed more evaluation. A retrospective study reported that osimertinib after nivolumab increased the frequency of hepatotoxicity in NSCLC patients [134]. A phase II clinical trial (NCT03082534) reported that cetuximab plus pembrolizumab showed promising activity for advanced HNSCC patients [135]. A phase Ib/II study (NCT02713373) showed that cetuximab plus pembrolizumab was well tolerated in CRC patients [136]. In a phase II trial (NCT03442569), the combination regimen of panitumumab, ipilimumab, and nivolumab showed a good median PFS (5.7 months) [137]. In a phase II study in advanced NSCLC patients, necitumumab and pembrolizumab showed potential benefits (median PFS = 4.1 months) with no additive side effects as second-line therapy [138]. According to the present evidence, ICIs combined with EGFR TKIs may cause more adverse effects, while ICIs and EGFR mAbs are promising treatments with favorable benefits and controllable toxicity (Table 1).
Approved HER2 inhibitors include neratinib, tucatinib, afatinib, pertuzumab, and trastuzumab, which target the overexpression of HER2 receptors in NSCLC, breast cancer, and gastric cancer [139]. In the phase II ALPHA clinical trial (NCT03695510), afatinib plus pembrolizumab demonstrated potential benefits for advanced HNSCC patients, with 53.8% of patients showing an objective response [140]. In the phase Ib/II PANACEA trial (NCT02129556), the addition of pembrolizumab brought durable clinical benefit to trastuzumab-resistant HER2+ breast cancer patients [141]. Pembrolizumab plus trastuzumab also showed a similar trend as the first-line treatment of esophageal, gastric, or gastro-esophageal junction cancer (NCT02954536) [142]. However, these results did not include control groups, and further trials are required to assess the efficacy and safety compared with current therapies (Table 2).

4.4. PARP Inhibitors with ICIs

PARP plays a vital role in DNA repair pathways, and tumors with defective homologous recombination, especially BRCA-deficient, are susceptible to PARP inhibitors, including olaparib, talazoparib, niraparib, and rucaparib [143]. In a phase I/II clinical trial for recurrent platinum-resistant ovarian cancer (NCT02657889), niraparib plus pembrolizumab showed promising antitumor activity with an ORR of 18% [144]. A phase II clinical trial that investigated the combination of olaparib and durvalumab in advanced prostate and ovarian cancer reported modest efficacy with an acceptable safety profile [145,146]. Several other phase II studies are still ongoing [147,148]. Although there is a long way to go before clinical use, the combination of PARP inhibitors and ICIs may be of great value for patients with specific mutations.

4.5. FGFR/PDGFR/VEGFR Signaling Inhibitors with ICIs

The activation of receptor tyrosine kinases, such as FGFR, PDGFR, and VEGFR, is involved in the development and metastasis of various human cancers [149]. Inhibitors of these kinases have been widely applied in treating HCC, RCC, and differentiated thyroid cancer (DTC). A phase Ib clinical trial (NCT03628521) showed the encouraging efficacy (median PFS = 15 months; ORR = 72.7%) of anlotinib plus sintilimab, an anti-PD-1 antibody, with tolerable adverse events in NSCLC patients [150]. Recent phase II trials also reported the efficacy and safety of apatinib plus camrelizumab, an anti-PD-1 antibody, in advanced TNBC (NCT03394287) and HCC (NCT02942329) [151,152]. However, apatinib plus camrelizumab failed to improve PFS and ORR in metastatic CRC in a phase II trial (NCT03912857) [153]. In the phase III KEYNOTE-426 trial (NCT02853331), axitinib plus pembrolizumab showed longer PFS (15.4 vs. 11.1 months, respectively; p < 0.0001) than sunitinib monotherapy as a first-line treatment for advanced RCC [154]. Cabozantinib plus nivolumab showed longer PFS (16.6 vs. 8.3 months, respectively; p < 0.001) and higher ORR (55.7% vs. 27.1%, respectively; p < 0.001) than sunitinib monotherapy as a first-line treatment for advance RCC [155]. Lenvatinib combined with pembrolizumab showed encouraging clinical benefits in PFS, OS, and ORR with manageable toxicity in advanced papillary thyroid carcinoma (PTC), HCC, endometrial cancer, RCC, and gastric cancer according to recent clinical trials [156,157,158,159,160]. In a phase II study, metastatic CRC patients receiving regorafenib plus avelumab showed a median OS of 10.8 months and a median PFS of 3.6 months without unexpected side effects [161]. In a retrospective study, regorafenib plus sintilimab showed better OS (13.4 vs. 9.9 months; p = 0.023), longer PFS (5.6 vs. 4.0 months; p = 0.045), and higher ORR (36.2% vs. 16.4%; p = 0.045) than regorafenib alone as a second-line treatment for advance HCC [162]. Pemigatnib, a selective FGFR inhibitor, showed tolerable toxicity and potential antitumor activity in combination with pembrolizumab for advanced cancer based on the phase I/II FIGHT-101 trial (NCT02393248) [163]. Retrospective studies also revealed that TKIs plus anti-PD-1 antibodies, including nivolumab, pembrolizumab, or sintilimab, showed potential benefits with tolerable toxicity in advanced HCC patients [164]. These results suggested the encouraging efficacy and safety of multi-targeted TKIs combined with ICIs (Table 2).
Bevacizumab works as an antitumor agent in clinical via its blockade of VEGF. Bevacizumab, together with atezolizumab, is recommended as first-line therapy for advanced HCC because the combination showed better OS (64.2% vs. 57.6% at 12 months) and PFS (6.8 vs. 4.3 months; p < 0.001) than sorafenib in the phase III trial (NCT03434379) [165]. This combination also improved PFS (11.2 vs. 7.7 months) than sunitinib for advanced RCC patients in the phase III clinical trial (NCT02420821) [166]. Sintilimab, another VEGF mAb, showed improved PFS (6.9 vs. 4.3 months) combined with sintilimab and chemotherapy than chemotherapy alone for NSCLC patients in the phase III study. These results indicated the potential efficacy with good tolerance of VEGF mAb plus ICIs as first-line therapies (Table 2). To conclude, the application of FGFR/PDGFR/VEGFR signaling inhibitors plus ICIs may provide promising benefits with similar adverse events for patients with malignancies.

4.6. Epigenetic Agents with ICIs

Epigenetic agents are emerging combination partners for the treatment of malignancies, and recent studies investigated their role in combination with ICIs (Table 3). Entinostat, an HDAC inhibitor, demonstrated promising clinical benefits with an ORR of 9.2% in NSCLC and 14% in metastatic uveal melanoma in combination with pembrolizumab in phase II clinical trials [167,168]. Azacitidine, a DNMT inhibitor approved for acute myeloid leukemia (AML), showed encouraging clinical activity in combination with nivolumab for AML patients but limited efficacy with avelumab (ORR = 10.5%) [169,170]. Decitabine, another DNMT inhibitor, achieved bot ah high response rate (ORR = 52%) and long-term survival improvement (median PFS = 20.0 months) with camrelizumab [169]. These results indicated that anti-tumor activity with acceptable toxicity might be possible with epigenetic agents plus ICIs, especially anti-PD-1 antibodies.

5. Discussion

In addition to blocking cell growth, targeted therapy contributes to the TIME remodeling that enhances the anti-tumor response. Treatment response to ICIs is mainly dependent on an active TIME. Therefore, targeted therapy can be a potent option to improve the efficacy of ICIs, possibly through mechanisms including the reinforcement of effector T cell infiltration and the impairment of immunosuppressive cells. Indeed, the strategy is being investigated in many clinical trials, but most of them are in an early stage (Table 1, Table 2 and Table 3). To optimize the combinational therapeutic approach, several issues are noteworthy.
The first is how the proteins or signaling pathways interfered with by targeted agents influence each immune component in TIME and their integrated effect on the antitumor response. Corresponding studies have been conducted in preclinical models and patient cohorts. As an example, VEGF signaling inhibitors, demonstrated to activate effector T cells, promote DC maturation, and boost Treg cell depletion, were approved for treating various cancers in combination with ICIs [27,171]. However, some agents modulate an immunosuppressive TIME by impairing effector immune cells’ function, enhancing the recruitment of immunosuppressive cells and promoting polarization to pro-tumor phenotype [34,80].
The second is whether the increasing anti-tumor activities of the combination strategy escalate the toxicities. The side effects induced by molecularly targeted agents are attributed to the inhibition of targets in normal tissues, such as rash, hypertension, and hepatoxicity. Immune checkpoints protect the body from damage as a consequence of dysregulated immunity. Therefore, the immune-related adverse events (irAEs) coupled with ICIs may be due to an imbalance in immunologic homeostasis. Assessing the treatment safety and finding biomarkers of side effects is important.
Selecting appropriate patient subgroups for combined treatment and discovering potential biomarkers to predict the safety and efficacy are essential. Without synergy effects of the drug combination, choosing patient subgroups precisely can also contribute to the improved response of combination therapy [172]. Thus, beyond molecular features of cancers, more elements that were previously ignored are being integrated into the criteria for patient subgroups, such as age, sex, and lifestyle choices. Observation based on age strata showed significant differences in cancer biology and immune functions in older vs. younger patients [173]. The published reports illustrated that ICIs are more effective in the elderly, which may be due to the upregulated expression of immune checkpoints with age [174]. However, the physiological changes in the elderly may influence the pharmacology of anticancer drugs. As an example, in patients older than 75 years, CDK4/6 inhibitors induced higher rates of adverse effects, which decreased the quality of life [175]. These studies informed clinicians about making tailored therapeutic strategies for each patient subgroup, maximizing efficacy and minimizing toxicity.
Despite ICIs, novel immunotherapy approaches such as cellular therapy and cancer vaccine are emerging. Adoptive cellular therapy (ACT) has been approved to treat hematologic malignancies, but its exploitation is hindered in solid tumors. One of the barriers is immunosuppressive TIME. Applying targeted agents to directly eliminate or reprogram immunosuppressive cells is a promising approach to improving the efficacy of ACT. For instance, CDK4/6 inhibitors reduce the proportion of MDSC and Treg cells in patients with metastatic breast cancer. Therefore, using targeted agents simultaneously with or before ACT may enhance the antitumor response.
Reusing molecularly targeted agents as an adjuvant in immunotherapy is a field that merits further study. Taken together, targeted therapy in combination with immunotherapies has shown great potential, which can lead to superior clinical efficacy.

Funding

This work was supported by the National Natural Science Foundation of China (No. 82130077) and the Research Projects from the Science and Technology Commission of Shanghai Municipality (Grants 21JC1410100, 21JC1401200). The study sponsor did not participate in the study design, collection, analysis, or interpretation of data.

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Data Availability Statement

All data supporting reported results can be found in this manuscript.

Conflicts of Interest

The authors declare no conflict of interest.

Abbreviations

ADCCAntibody-Dependent Cell-Mediated Cytotoxicity
AMLAcute Myeloid Leukemia
BTKBruton Tyrosine Kinase
CDKCycle-dependent Kinase
CLLChronic Lymphatic Leukemia
CRCColorectal Cancer
DCDendritic Cells
DDRDNA Damage Repair
DNMTDNA Methyltransferase
DTCDifferentiated Thyroid Cancer
HCCHepatocellular Carcinoma
HDACHistone Deacetylases
HNSCCHead and Neck Squamous Cell Carcinoma
ICIImmune Checkpoint Inhibitor
MCLMantel Cell Lymphoma
MDSCMyeloid Derived Suppressor Cells
NKNature Killer Cells
NSCLCNon-small Cell Lung Cancer
ORRObjective Response Rate
OSOverall Survival
PDACPancreatic Ductal Adenocarcinoma
PFSProgression-free Survival
PTCPapillary Thyroid Carcinoma
RCCRenal Cell Carcinoma
TAMTumor-associated Macrophages
TGF-βtransforming growth factor-β
TIBTumor-infiltrating B Cells
TIMETumor Immune Microenvironment
TKITyrosine Kinase Inhibitor
TNBCTriple-negative Breast Cancer

References

  1. Sung, H.; Ferlay, J.; Siegel, R.L.; Laversanne, M.; Soerjomataram, I.; Jemal, A.; Bray, F. Global Cancer Statistics 2020: GLOBOCAN Estimates of Incidence and Mortality Worldwide for 36 Cancers in 185 Countries. CA A Cancer J. Clin. 2021, 71, 209–249. [Google Scholar] [CrossRef] [PubMed]
  2. Falzone, L.; Salomone, S.; Libra, M. Evolution of Cancer Pharmacological Treatments at the Turn of the Third Millennium. Front Pharm. 2018, 9, 1300. [Google Scholar] [CrossRef] [PubMed]
  3. Tsimberidou, A.-M. Targeted therapy in cancer. Cancer Chemother. Pharmacol. 2015, 76, 1113–1132. [Google Scholar] [CrossRef] [PubMed]
  4. Folkman, J. Tumor Angiogenesis: Therapeutic Implications. N. Engl. J. Med. 1971, 285, 1182–1186. [Google Scholar] [CrossRef]
  5. Keefe, D.M.K.; Bateman, E.H. Potential Successes and Challenges of Targeted Cancer Therapies. J. Natl. Cancer Inst. Monogr. 2019, 2019, lgz008. [Google Scholar] [CrossRef]
  6. Petroni, G.; Buque, A.; Zitvogel, L.; Kroemer, G.; Galluzzi, L. Immunomodulation by targeted anticancer agents. Cancer Cell 2021, 39, 310–345. [Google Scholar] [CrossRef]
  7. Tao, L.; Zhu, F.; Xu, F.; Chen, Z.; Jiang, Y.Y.; Chen, Y.Z. Co-targeting cancer drug escape pathways confers clinical advantage for multi-target anticancer drugs. Pharm. Res. 2015, 102, 123–131. [Google Scholar] [CrossRef]
  8. Gonzalez-Aparicio, M.; Alfaro, C. Implication of Interleukin Family in Cancer Pathogenesis and Treatment. Cancers 2021, 13, 1016. [Google Scholar] [CrossRef]
  9. Yang, L.; Pang, Y.; Moses, H.L. TGF-beta and immune cells: An important regulatory axis in the tumor microenvironment and progression. Trends Immunol. 2010, 31, 220–227. [Google Scholar] [CrossRef]
  10. Bagchi, S.; Yuan, R.; Engleman, E.G. Immune Checkpoint Inhibitors for the Treatment of Cancer: Clinical Impact and Mechanisms of Response and Resistance. Annu. Rev. Pathol. 2021, 16, 223–249. [Google Scholar] [CrossRef]
  11. Tawbi, H.A.; Schadendorf, D.; Lipson, E.J.; Ascierto, P.A.; Matamala, L.; Castillo Gutierrez, E.; Rutkowski, P.; Gogas, H.J.; Lao, C.D.; De Menezes, J.J.; et al. Relatlimab and Nivolumab versus Nivolumab in Untreated Advanced Melanoma. N. Engl. J. Med. 2022, 386, 24–34. [Google Scholar] [CrossRef] [PubMed]
  12. Kohlhapp, F.J.; Haribhai, D.; Mathew, R.; Duggan, R.; Ellis, P.A.; Wang, R.; Lasater, E.A.; Shi, Y.; Dave, N.; Riehm, J.J.; et al. Venetoclax Increases Intratumoral Effector T Cells and Antitumor Efficacy in Combination with Immune Checkpoint Blockade. Cancer Discov. 2021, 11, 68–79. [Google Scholar] [CrossRef] [PubMed]
  13. Cheng, A.-L.; Qin, S.; Ikeda, M.; Galle, P.; Ducreux, M.; Zhu, A.; Kim, T.-Y.; Kudo, M.; Breder, V.; Merle, P. IMbrave150: Efficacy and safety results from a ph III study evaluating atezolizumab (atezo)+ bevacizumab (bev) vs sorafenib (Sor) as first treatment (tx) for patients (pts) with unresectable hepatocellular carcinoma (HCC). Ann. Oncol. 2019, 30, ix186–ix187. [Google Scholar] [CrossRef]
  14. Finn, R.S.; Ryoo, B.Y.; Merle, P.; Kudo, M.; Bouattour, M.; Lim, H.Y.; Breder, V.; Edeline, J.; Chao, Y.; Ogasawara, S.; et al. Pembrolizumab As Second-Line Therapy in Patients With Advanced Hepatocellular Carcinoma in KEYNOTE-240: A Randomized, Double-Blind, Phase III Trial. J. Clin. Oncol. 2020, 38, 193–202. [Google Scholar] [CrossRef]
  15. El Bairi, K.; Haynes, H.R.; Blackley, E.; Fineberg, S.; Shear, J.; Turner, S.; de Freitas, J.R.; Sur, D.; Amendola, L.C.; Gharib, M.; et al. The tale of TILs in breast cancer: A report from The International Immuno-Oncology Biomarker Working Group. NPJ Breast Cancer 2021, 7, 150. [Google Scholar] [CrossRef]
  16. 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, 5, 1431–1438. [Google Scholar] [CrossRef]
  17. Leone, R.D.; Powell, J.D. Metabolism of immune cells in cancer. Nat. Rev. Cancer 2020, 20, 516–531. [Google Scholar] [CrossRef]
  18. Deng, J.; Wang, E.S.; Jenkins, R.W.; Li, S.; Dries, R.; Yates, K.; Chhabra, S.; Huang, W.; Liu, H.; Aref, A.R.; et al. CDK4/6 Inhibition Augments Antitumor Immunity by Enhancing T-cell Activation. Cancer Discov. 2018, 8, 216–233. [Google Scholar] [CrossRef]
  19. Liu, C.; Peng, W.; Xu, C.; Lou, Y.; Zhang, M.; Wargo, J.A.; Chen, J.Q.; Li, H.S.; Watowich, S.S.; Yang, Y.; et al. BRAF inhibition increases tumor infiltration by T cells and enhances the antitumor activity of adoptive immunotherapy in mice. Clin. Cancer Res. 2013, 19, 393–403. [Google Scholar] [CrossRef]
  20. Donia, M.; Fagone, P.; Nicoletti, F.; Andersen, R.S.; Hogdall, E.; Straten, P.T.; Andersen, M.H.; Svane, I.M. BRAF inhibition improves tumor recognition by the immune system: Potential implications for combinatorial therapies against melanoma involving adoptive T-cell transfer. Oncoimmunology 2012, 1, 1476–1483. [Google Scholar] [CrossRef] [Green Version]
  21. Long, G.V.; Hauschild, A.; Santinami, M.; Atkinson, V.; Mandala, M.; Chiarion-Sileni, V.; Larkin, J.; Nyakas, M.; Dutriaux, C.; Haydon, A.; et al. Adjuvant Dabrafenib plus Trametinib in Stage III BRAF-Mutated Melanoma. N. Engl. J. Med. 2017, 377, 1813–1823. [Google Scholar] [CrossRef] [PubMed]
  22. Erkes, D.A.; Cai, W.; Sanchez, I.M.; Purwin, T.J.; Rogers, C.; Field, C.O.; Berger, A.C.; Hartsough, E.J.; Rodeck, U.; Alnemri, E.S.; et al. Mutant BRAF and MEK Inhibitors Regulate the Tumor Immune Microenvironment via Pyroptosis. Cancer Discov. 2020, 10, 254–269. [Google Scholar] [CrossRef] [PubMed]
  23. Peiffer, L.; Farahpour, F.; Sriram, A.; Spassova, I.; Hoffmann, D.; Kubat, L.; Stoitzner, P.; Gambichler, T.; Sucker, A.; Ugurel, S.; et al. BRAF and MEK inhibition in melanoma patients enables reprogramming of tumor infiltrating lymphocytes. Cancer Immunol. Immunother. 2021, 70, 1635–1647. [Google Scholar] [CrossRef] [PubMed]
  24. Hoyer, S.; Eberlein, V.; Schuler, G.; Berking, C.; Heinzerling, L.; Schaft, N.; Dorrie, J. BRAF and MEK Inhibitors Affect Dendritic-Cell Maturation and T-Cell Stimulation. Int. J. Mol. Sci. 2021, 22, 1951. [Google Scholar] [CrossRef] [PubMed]
  25. Zhou, X.; Singh, M.; Sanz Santos, G.; Guerlavais, V.; Carvajal, L.A.; Aivado, M.; Zhan, Y.; Oliveira, M.M.S.; Westerberg, L.S.; Annis, D.A.; et al. Pharmacologic Activation of p53 Triggers Viral Mimicry Response Thereby Abolishing Tumor Immune Evasion and Promoting Antitumor Immunity. Cancer Discov. 2021, 11, 3090–3105. [Google Scholar] [CrossRef]
  26. Voron, T.; Colussi, O.; Marcheteau, E.; Pernot, S.; Nizard, M.; Pointet, A.L.; Latreche, S.; Bergaya, S.; Benhamouda, N.; Tanchot, C.; et al. VEGF-A modulates expression of inhibitory checkpoints on CD8+ T cells in tumors. J. Exp. Med. 2015, 212, 139–148. [Google Scholar] [CrossRef]
  27. Xue, L.; Gao, X.; Zhang, H.; Tang, J.; Wang, Q.; Li, F.; Li, X.; Yu, X.; Lu, Z.; Huang, Y.; et al. Antiangiogenic antibody BD0801 combined with immune checkpoint inhibitors achieves synergistic antitumor activity and affects the tumor microenvironment. BMC Cancer 2021, 21, 1134. [Google Scholar] [CrossRef]
  28. Llovet, J.M.; Kelley, R.K.; Villanueva, A.; Singal, A.G.; Pikarsky, E.; Roayaie, S.; Lencioni, R.; Koike, K.; Zucman-Rossi, J.; Finn, R.S. Hepatocellular carcinoma. Nat. Rev. Dis. Primers 2021, 7, 6. [Google Scholar] [CrossRef]
  29. Chen, M.L.; Yan, B.S.; Lu, W.C.; Chen, M.H.; Yu, S.L.; Yang, P.C.; Cheng, A.L. Sorafenib relieves cell-intrinsic and cell-extrinsic inhibitions of effector T cells in tumor microenvironment to augment antitumor immunity. Int. J. Cancer 2014, 134, 319–331. [Google Scholar] [CrossRef]
  30. Kalathil, S.G.; Hutson, A.; Barbi, J.; Iyer, R.; Thanavala, Y. Augmentation of IFN-gamma+ CD8+ T cell responses correlates with survival of HCC patients on sorafenib therapy. JCI Insight 2019, 4, e130116. [Google Scholar] [CrossRef]
  31. Lu, M.; Zhang, X.; Gao, X.; Sun, S.; Wei, X.; Hu, X.; Huang, C.; Xu, H.; Wang, B.; Zhang, W.; et al. Lenvatinib enhances T cell immunity and the efficacy of adoptive chimeric antigen receptor-modified T cells by decreasing myeloid-derived suppressor cells in cancer. Pharm. Res 2021, 174, 105829. [Google Scholar] [CrossRef] [PubMed]
  32. 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] [PubMed]
  33. Jia, Y.; Li, X.; Jiang, T.; Zhao, S.; Zhao, C.; Zhang, L.; Liu, X.; Shi, J.; Qiao, M.; Luo, J.; et al. EGFR-targeted therapy alters the tumor microenvironment in EGFR-driven lung tumors: Implications for combination therapies. Int. J. Cancer 2019, 145, 1432–1444. [Google Scholar] [CrossRef] [PubMed]
  34. Moreno-Lama, L.; Galindo-Campos, M.A.; Martinez, C.; Comerma, L.; Vazquez, I.; Vernet-Tomas, M.; Ampurdanes, 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]
  35. Sugiyama, E.; Togashi, Y.; Takeuchi, Y.; Shinya, S.; Tada, Y.; Kataoka, K.; Tane, K.; Sato, E.; Ishii, G.; Goto, K.; et al. Blockade of EGFR improves responsiveness to PD-1 blockade in EGFR-mutated non-small cell lung cancer. Sci. Immunol. 2020, 5, eaav3937. [Google Scholar] [CrossRef] [PubMed]
  36. Sharabi, A.; Ghera, N.H. Breaking tolerance in a mouse model of multiple myeloma by chemoimmunotherapy. Adv. Cancer Res. 2010, 107, 1–37. [Google Scholar] [CrossRef] [PubMed]
  37. Ghiringhelli, F.; Larmonier, N.; Schmitt, E.; Parcellier, A.; Cathelin, D.; Garrido, C.; Chauffert, B.; Solary, E.; Bonnotte, B.; Martin, F. CD4+CD25+ regulatory T cells suppress tumor immunity but are sensitive to cyclophosphamide which allows immunotherapy of established tumors to be curative. Eur. J. Immunol. 2004, 34, 336–344. [Google Scholar] [CrossRef] [PubMed]
  38. Nizar, S.; Copier, J.; Meyer, B.; Bodman-Smith, M.; Galustian, C.; Kumar, D.; Dalgleish, A. T-regulatory cell modulation: The future of cancer immunotherapy? Br. J. Cancer 2009, 100, 1697–1703. [Google Scholar] [CrossRef]
  39. Goel, S.; DeCristo, M.J.; Watt, A.C.; BrinJones, H.; Sceneay, J.; Li, B.B.; Khan, N.; Ubellacker, J.M.; Xie, S.; Metzger-Filho, O.; et al. CDK4/6 inhibition triggers anti-tumour immunity. Nature 2017, 548, 471–475. [Google Scholar] [CrossRef]
  40. Peuker, C.A.; Yaghobramzi, S.; Grunert, C.; Keilholz, L.; Gjerga, E.; Hennig, S.; Schaper, S.; Na, I.K.; Keller, U.; Brucker, S.; et al. Treatment with ribociclib shows favourable immunomodulatory effects in patients with hormone receptor-positive breast cancer-findings from the RIBECCA trial. Eur. J. Cancer 2022, 162, 45–55. [Google Scholar] [CrossRef]
  41. Torrens, L.; Montironi, C.; Puigvehi, M.; Mesropian, A.; Leslie, J.; Haber, P.K.; Maeda, M.; Balaseviciute, U.; Willoughby, C.E.; Abril-Fornaguera, J.; et al. Immunomodulatory Effects of Lenvatinib Plus Anti-Programmed Cell Death Protein 1 in Mice and Rationale for Patient Enrichment in Hepatocellular Carcinoma. Hepatology 2021, 74, 2652–2669. [Google Scholar] [CrossRef]
  42. Isoyama, S.; Mori, S.; Sugiyama, D.; Kojima, Y.; Tada, Y.; Shitara, K.; Hinohara, K.; Dan, S.; Nishikawa, H. Cancer immunotherapy with PI3K and PD-1 dual-blockade via optimal modulation of T cell activation signal. J. Immunother. Cancer 2021, 9, e002279. [Google Scholar] [CrossRef]
  43. Eschweiler, S.; Ramirez-Suastegui, C.; Li, Y.; King, E.; Chudley, L.; Thomas, J.; Wood, O.; von Witzleben, A.; Jeffrey, D.; McCann, K.; et al. Intermittent PI3Kdelta inhibition sustains anti-tumour immunity and curbs irAEs. Nature 2022, 605, 741–746. [Google Scholar] [CrossRef]
  44. Shang, B.; Liu, Y.; Jiang, S.J.; Liu, Y. Prognostic value of tumor-infiltrating FoxP3+ regulatory T cells in cancers: A systematic review and meta-analysis. Sci. Rep. 2015, 5, 15179. [Google Scholar] [CrossRef]
  45. Frey, D.M.; Droeser, R.A.; Viehl, C.T.; Zlobec, I.; Lugli, A.; Zingg, U.; Oertli, D.; Kettelhack, C.; Terracciano, L.; Tornillo, L. High frequency of tumor-infiltrating FOXP3(+) regulatory T cells predicts improved survival in mismatch repair-proficient colorectal cancer patients. Int. J. Cancer 2010, 126, 2635–2643. [Google Scholar] [CrossRef]
  46. Lee, W.S.; Park, S.; Lee, W.Y.; Yun, S.H.; Chun, H.K. Clinical impact of tumor-infiltrating lymphocytes for survival in stage II colon cancer. Cancer 2010, 116, 5188–5199. [Google Scholar] [CrossRef]
  47. Downs-Canner, S.M.; Meier, J.; Vincent, B.G.; Serody, J.S. B Cell Function in the Tumor Microenvironment. Annu. Rev. Immunol. 2022, 40, 169–193. [Google Scholar] [CrossRef]
  48. Fridman, W.H.; Meylan, M.; Petitprez, F.; Sun, C.M.; Italiano, A.; Sautes-Fridman, C. B cells and tertiary lymphoid structures as determinants of tumour immune contexture and clinical outcome. Nat. Rev. Clin. Oncol. 2022, 19, 441–457. [Google Scholar] [CrossRef]
  49. Liang, C.; Tian, D.; Ren, X.; Ding, S.; Jia, M.; Xin, M.; Thareja, S. The development of Bruton’s tyrosine kinase (BTK) inhibitors from 2012 to 2017: A mini-review. Eur. J. Med. Chem. 2018, 151, 315–326. [Google Scholar] [CrossRef]
  50. Gunderson, A.J.; Kaneda, M.M.; Tsujikawa, T.; Nguyen, A.V.; Affara, N.I.; Ruffell, B.; Gorjestani, S.; Liudahl, S.M.; Truitt, M.; Olson, P.; et al. Bruton Tyrosine Kinase-Dependent Immune Cell Cross-talk Drives Pancreas Cancer. Cancer Discov. 2016, 6, 270–285. [Google Scholar] [CrossRef] [Green Version]
  51. Zhang, Q.F.; Li, J.; Jiang, K.; Wang, R.; Ge, J.L.; Yang, H.; Liu, S.J.; Jia, L.T.; Wang, L.; Chen, B.L. CDK4/6 inhibition promotes immune infiltration in ovarian cancer and synergizes with PD-1 blockade in a B cell-dependent manner. Theranostics 2020, 10, 10619–10633. [Google Scholar] [CrossRef]
  52. Helmink, B.A.; Reddy, S.M.; Gao, J.; Zhang, S.; Basar, R.; Thakur, R.; Yizhak, K.; Sade-Feldman, M.; Blando, J.; Han, G.; et al. B cells and tertiary lymphoid structures promote immunotherapy response. Nature 2020, 577, 549–555. [Google Scholar] [CrossRef]
  53. Cabrita, R.; Lauss, M.; Sanna, A.; Donia, M.; Skaarup Larsen, M.; Mitra, S.; Johansson, I.; Phung, B.; Harbst, K.; Vallon-Christersson, J.; et al. Tertiary lymphoid structures improve immunotherapy and survival in melanoma. Nature 2020, 577, 561–565. [Google Scholar] [CrossRef]
  54. Van Acker, H.H.; Capsomidis, A.; Smits, E.L.; Van Tendeloo, V.F. CD56 in the Immune System: More Than a Marker for Cytotoxicity? Front. Immunol. 2017, 8, 892. [Google Scholar] [CrossRef]
  55. Fu, B.; Tian, Z.; Wei, H. Subsets of human natural killer cells and their regulatory effects. Immunology 2014, 141, 483–489. [Google Scholar] [CrossRef]
  56. Wang, W.; Jiang, J.; Wu, C. CAR-NK for tumor immunotherapy: Clinical transformation and future prospects. Cancer Lett. 2020, 472, 175–180. [Google Scholar] [CrossRef]
  57. Ruscetti, M.; Leibold, J.; Bott, M.J.; Fennell, M.; Kulick, A.; Salgado, N.R.; Chen, C.C.; Ho, Y.J.; Sanchez-Rivera, F.J.; Feucht, J.; et al. NK cell-mediated cytotoxicity contributes to tumor control by a cytostatic drug combination. Science 2018, 362, 1416–1422. [Google Scholar] [CrossRef]
  58. Witalisz-Siepracka, A.; Gotthardt, D.; Prchal-Murphy, M.; Didara, Z.; Menzl, I.; Prinz, D.; Edlinger, L.; Putz, E.M.; Sexl, V. NK Cell-Specific CDK8 Deletion Enhances Antitumor Responses. Cancer Immunol. Res. 2018, 6, 458–466. [Google Scholar] [CrossRef]
  59. Ke, C.; Hou, H.; Li, J.; Su, K.; Huang, C.; Lin, Y.; Lu, Z.; Du, Z.; Tan, W.; Yuan, Z. Extracellular Vesicle Delivery of TRAIL Eradicates Resistant Tumor Growth in Combination with CDK Inhibition by Dinaciclib. Cancers 2020, 12, 1157. [Google Scholar] [CrossRef]
  60. Ferrari de Andrade, L.; Ngiow, S.F.; Stannard, K.; Rusakiewicz, S.; Kalimutho, M.; Khanna, K.K.; Tey, S.K.; Takeda, K.; Zitvogel, L.; Martinet, L.; et al. Natural killer cells are essential for the ability of BRAF inhibitors to control BRAFV600E-mutant metastatic melanoma. Cancer Res. 2014, 74, 7298–7308. [Google Scholar] [CrossRef] [Green Version]
  61. Mallmann-Gottschalk, N.; Sax, Y.; Kimmig, R.; Lang, S.; Brandau, S. EGFR-Specific Tyrosine Kinase Inhibitor Modifies NK Cell-Mediated Antitumoral Activity against Ovarian Cancer Cells. Int. J. Mol. Sci. 2019, 20, 4693. [Google Scholar] [CrossRef]
  62. Lopez-Cobo, S.; Pieper, N.; Campos-Silva, C.; Garcia-Cuesta, E.M.; Reyburn, H.T.; Paschen, A.; Vales-Gomez, M. Impaired NK cell recognition of vemurafenib-treated melanoma cells is overcome by simultaneous application of histone deacetylase inhibitors. Oncoimmunology 2018, 7, e1392426. [Google Scholar] [CrossRef]
  63. Rohrbacher, L.; Brauchle, B.; Ogrinc Wagner, A.; von Bergwelt-Baildon, M.; Bucklein, V.L.; Subklewe, M. The PI3K partial differential-Selective Inhibitor Idelalisib Induces T- and NK-Cell Dysfunction Independently of B-Cell Malignancy-Associated Immunosuppression. Front. Immunol. 2021, 12, 608625. [Google Scholar] [CrossRef]
  64. Rossi, L.E.; Avila, D.E.; Spallanzani, R.G.; Ziblat, A.; Fuertes, M.B.; Lapyckyj, L.; Croci, D.O.; Rabinovich, G.A.; Domaica, C.I.; Zwirner, N.W. Histone deacetylase inhibitors impair NK cell viability and effector functions through inhibition of activation and receptor expression. J. Leukoc. Biol. 2012, 91, 321–331. [Google Scholar] [CrossRef]
  65. Powell, D.R.; Huttenlocher, A. Neutrophils in the Tumor Microenvironment. Trends Immunol. 2016, 37, 41–52. [Google Scholar] [CrossRef]
  66. Li, Y.; Hu, Q.; Li, W.; Liu, S.; Li, K.; Li, X.; Du, J.; Yu, Z.; Wang, C.; Zhang, C. Simultaneous blockage of contextual TGF-beta by cyto-pharmaceuticals to suppress breast cancer metastasis. J. Control. Release 2021, 336, 40–53. [Google Scholar] [CrossRef]
  67. Peng, H.; Shen, J.; Long, X.; Zhou, X.; Zhang, J.; Xu, X.; Huang, T.; Xu, H.; Sun, S.; Li, C.; et al. Local Release of TGF-beta Inhibitor Modulates Tumor-Associated Neutrophils and Enhances Pancreatic Cancer Response to Combined Irreversible Electroporation and Immunotherapy. Adv. Sci. 2022, 9, e2105240. [Google Scholar] [CrossRef]
  68. Harizi, H. Reciprocal crosstalk between dendritic cells and natural killer cells under the effects of PGE2 in immunity and immunopathology. Cell. Mol. Immunol. 2013, 10, 213–221. [Google Scholar] [CrossRef]
  69. Shang, K.; Bai, Y.P.; Wang, C.; Wang, Z.; Gu, H.Y.; Du, X.; Zhou, X.Y.; Zheng, C.L.; Chi, Y.Y.; Mukaida, N.; et al. Crucial involvement of tumor-associated neutrophils in the regulation of chronic colitis-associated carcinogenesis in mice. PLoS ONE 2012, 7, e51848. [Google Scholar] [CrossRef]
  70. Ning, C.; Li, Y.Y.; Wang, Y.; Han, G.C.; Wang, R.X.; Xiao, H.; Li, X.Y.; Hou, C.M.; Ma, Y.F.; Sheng, D.S.; et al. Complement activation promotes colitis-associated carcinogenesis through activating intestinal IL-1beta/IL-17A axis. Mucosal. Immunol. 2015, 8, 1275–1284. [Google Scholar] [CrossRef] [Green Version]
  71. Carreno, B.M.; Magrini, V.; Becker-Hapak, M.; Kaabinejadian, S.; Hundal, J.; Petti, A.A.; Ly, A.; Lie, W.R.; Hildebrand, W.H.; Mardis, E.R.; et al. Cancer immunotherapy. A dendritic cell vaccine increases the breadth and diversity of melanoma neoantigen-specific T cells. Science 2015, 348, 803–808. [Google Scholar] [CrossRef]
  72. Gall, V.A.; Philips, A.V.; Qiao, N.; Clise-Dwyer, K.; Perakis, A.A.; Zhang, M.; Clifton, G.T.; Sukhumalchandra, P.; Ma, Q.; Reddy, S.M.; et al. Trastuzumab Increases HER2 Uptake and Cross-Presentation by Dendritic Cells. Cancer Res 2017, 77, 5374–5383. [Google Scholar] [CrossRef]
  73. Aksoy, E.; Saveanu, L.; Manoury, B. The Isoform Selective Roles of PI3Ks in Dendritic Cell Biology and Function. Front. Immunol. 2018, 9, 2574. [Google Scholar] [CrossRef]
  74. Zhu, S.; Ma, A.H.; Zhu, Z.; Adib, E.; Rao, T.; Li, N.; Ni, K.; Chittepu, V.; Prabhala, R.; Garisto Risco, J.; et al. Synergistic antitumor activity of pan-PI3K inhibition and immune checkpoint blockade in bladder cancer. J. Immunother. Cancer 2021, 9, e002917. [Google Scholar] [CrossRef]
  75. Qian, B.Z.; Pollard, J.W. Macrophage diversity enhances tumor progression and metastasis. Cell 2010, 141, 39–51. [Google Scholar] [CrossRef]
  76. De Palma, M.; Lewis, C.E. Macrophage regulation of tumor responses to anticancer therapies. Cancer Cell 2013, 23, 277–286. [Google Scholar] [CrossRef]
  77. Poon, E.; Mullins, S.; Watkins, A.; Williams, G.S.; Koopmann, J.O.; Di Genova, G.; Cumberbatch, M.; Veldman-Jones, M.; Grosskurth, S.E.; Sah, V.; et al. The MEK inhibitor selumetinib complements CTLA-4 blockade by reprogramming the tumor immune microenvironment. J. Immunother. Cancer 2017, 5, 63. [Google Scholar] [CrossRef]
  78. Kaneda, M.M.; Cappello, P.; Nguyen, A.V.; Ralainirina, N.; Hardamon, C.R.; Foubert, P.; Schmid, M.C.; Sun, P.; Mose, E.; Bouvet, M.; et al. Macrophage PI3Kgamma Drives Pancreatic Ductal Adenocarcinoma Progression. Cancer Discov. 2016, 6, 870–885. [Google Scholar] [CrossRef]
  79. Fang, D.D.; Tang, Q.; Kong, Y.; Wang, Q.; Gu, J.; Fang, X.; Zou, P.; Rong, T.; Wang, J.; Yang, D.; et al. MDM2 inhibitor APG-115 synergizes with PD-1 blockade through enhancing antitumor immunity in the tumor microenvironment. J. Immunother. Cancer 2019, 7, 327. [Google Scholar] [CrossRef]
  80. Pander, J.; Heusinkveld, M.; van der Straaten, T.; Jordanova, E.S.; Baak-Pablo, R.; Gelderblom, H.; Morreau, H.; van der Burg, S.H.; Guchelaar, H.J.; van Hall, T. Activation of tumor-promoting type 2 macrophages by EGFR-targeting antibody cetuximab. Clin. Cancer Res. 2011, 17, 5668–5673. [Google Scholar] [CrossRef] [Green Version]
  81. 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] [PubMed]
  82. De Sanctis, F.; Solito, S.; Ugel, S.; Molon, B.; Bronte, V.; Marigo, I. MDSCs in cancer: Conceiving new prognostic and therapeutic targets. Biochim. Biophys. Acta 2016, 1865, 35–48. [Google Scholar] [CrossRef] [PubMed]
  83. Teo, Z.L.; Versaci, S.; Dushyanthen, S.; Caramia, F.; Savas, P.; Mintoff, C.P.; Zethoven, M.; Virassamy, B.; Luen, S.J.; McArthur, G.A.; et al. Combined CDK4/6 and PI3Kalpha Inhibition Is Synergistic and Immunogenic in Triple-Negative Breast Cancer. Cancer Res. 2017, 77, 6340–6352. [Google Scholar] [CrossRef] [PubMed]
  84. Allegrezza, M.J.; Rutkowski, M.R.; Stephen, T.L.; Svoronos, N.; Perales-Puchalt, A.; Nguyen, J.M.; Payne, K.K.; Singhal, S.; Eruslanov, E.B.; Tchou, J.; et al. Trametinib Drives T-cell-Dependent Control of KRAS-Mutated Tumors by Inhibiting Pathological Myelopoiesis. Cancer Res. 2016, 76, 6253–6265. [Google Scholar] [CrossRef]
  85. Lelliott, E.J.; Mangiola, S.; Ramsbottom, K.M.; Zethoven, M.; Lim, L.; Lau, P.K.H.; Oliver, A.J.; Martelotto, L.G.; Kirby, L.; Martin, C.; et al. Combined BRAF, MEK, and CDK4/6 Inhibition Depletes Intratumoral Immune-Potentiating Myeloid Populations in Melanoma. Cancer Immunol. Res. 2021, 9, 136–146. [Google Scholar] [CrossRef]
  86. Koinis, F.; Vetsika, E.K.; Aggouraki, D.; Skalidaki, E.; Koutoulaki, A.; Gkioulmpasani, M.; Georgoulias, V.; Kotsakis, A. Effect of First-Line Treatment on Myeloid-Derived Suppressor Cells’ Subpopulations in the Peripheral Blood of Patients with Non-Small Cell Lung Cancer. J. Thorac. Oncol. 2016, 11, 1263–1272. [Google Scholar] [CrossRef]
  87. Du Four, S.; Maenhout, S.K.; De Pierre, K.; Renmans, D.; Niclou, S.P.; Thielemans, K.; Neyns, B.; Aerts, J.L. Axitinib increases the infiltration of immune cells and reduces the suppressive capacity of monocytic MDSCs in an intracranial mouse melanoma model. Oncoimmunology 2015, 4, e998107. [Google Scholar] [CrossRef]
  88. Zhang, J.; Bu, X.; Wang, H.; Zhu, Y.; Geng, Y.; Nihira, N.T.; Tan, Y.; Ci, Y.; Wu, F.; Dai, X.; et al. Cyclin D-CDK4 kinase destabilizes PD-L1 via cullin 3-SPOP to control cancer immune surveillance. Nature 2018, 553, 91–95. [Google Scholar] [CrossRef]
  89. 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]
  90. 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]
  91. Sen, T.; Rodriguez, B.L.; Chen, L.; Corte, C.M.D.; 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]
  92. Chabanon, R.M.; Muirhead, G.; Krastev, D.B.; Adam, J.; Morel, D.; Garrido, M.; Lamb, A.; Henon, 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]
  93. Sato, H.; Niimi, A.; Yasuhara, T.; Permata, T.B.M.; Hagiwara, Y.; Isono, M.; Nuryadi, E.; Sekine, R.; Oike, T.; Kakoti, S.; et al. DNA double-strand break repair pathway regulates PD-L1 expression in cancer cells. Nat. Commun. 2017, 8, 1751. [Google Scholar] [CrossRef] [PubMed]
  94. Menzies, A.M.; Long, G.V. Dabrafenib and trametinib, alone and in combination for BRAF-mutant metastatic melanoma. Clin. Cancer Res. 2014, 20, 2035–2043. [Google Scholar] [CrossRef] [PubMed]
  95. Liu, L.; Mayes, P.A.; Eastman, S.; Shi, H.; Yadavilli, S.; Zhang, T.; Yang, J.; Seestaller-Wehr, L.; Zhang, S.Y.; Hopson, C.; et al. The BRAF and MEK Inhibitors Dabrafenib and Trametinib: Effects on Immune Function and in Combination with Immunomodulatory Antibodies Targeting PD-1, PD-L1, and CTLA-4. Clin. Cancer Res. 2015, 21, 1639–1651. [Google Scholar] [CrossRef] [PubMed]
  96. Stutvoet, T.S.; Kol, A.; de Vries, E.G.; de Bruyn, M.; Fehrmann, R.S.; Terwisscha van Scheltinga, A.G.; de Jong, S. MAPK pathway activity plays a key role in PD-L1 expression of lung adenocarcinoma cells. J Pathol 2019, 249, 52–64. [Google Scholar] [CrossRef]
  97. Xiong, W.; Zhang, B.; Yu, H.; Zhu, L.; Yi, L.; Jin, X. RRM2 Regulates Sensitivity to Sunitinib and PD-1 Blockade in Renal Cancer by Stabilizing ANXA1 and Activating the AKT Pathway. Adv. Sci. 2021, 8, e2100881. [Google Scholar] [CrossRef]
  98. Stadler, W.M.; Desai, A.A.; Quinn, D.I.; Bukowski, R.; Poiesz, B.; Kardinal, C.G.; Lewis, N.; Makalinao, A.; Murray, P.; Torti, F.M. A Phase I/II study of GTI-2040 and capecitabine in patients with renal cell carcinoma. Cancer Chemother. Pharm. 2008, 61, 689–694. [Google Scholar] [CrossRef]
  99. Zuckerman, J.E.; Gritli, I.; Tolcher, A.; Heidel, J.D.; Lim, D.; Morgan, R.; Chmielowski, B.; Ribas, A.; Davis, M.E.; Yen, Y. Correlating animal and human phase Ia/Ib clinical data with CALAA-01, a targeted, polymer-based nanoparticle containing siRNA. Proc. Natl. Acad. Sci. USA 2014, 111, 11449–11454. [Google Scholar] [CrossRef]
  100. Bouattour, M.; Raymond, E.; Qin, S.; Cheng, A.L.; Stammberger, U.; Locatelli, G.; Faivre, S. Recent developments of c-Met as a therapeutic target in hepatocellular carcinoma. Hepatology 2018, 67, 1132–1149. [Google Scholar] [CrossRef]
  101. Boccaccio, C.; Comoglio, P.M. Invasive growth: A MET-driven genetic programme for cancer and stem cells. Nat. Rev. Cancer 2006, 6, 637–645. [Google Scholar] [CrossRef] [PubMed]
  102. Rimassa, L.; Assenat, E.; Peck-Radosavljevic, M.; Pracht, M.; Zagonel, V.; Mathurin, P.; Rota Caremoli, E.; Porta, C.; Daniele, B.; Bolondi, L.; et al. Tivantinib for second-line treatment of MET-high, advanced hepatocellular carcinoma (METIV-HCC): A final analysis of a phase 3, randomised, placebo-controlled study. Lancet Oncol. 2018, 19, 682–693. [Google Scholar] [CrossRef]
  103. Li, H.; Li, C.W.; Li, X.; Ding, Q.; Guo, L.; Liu, S.; Liu, C.; Lai, C.C.; Hsu, J.M.; Dong, Q.; et al. MET Inhibitors Promote Liver Tumor Evasion of the Immune Response by Stabilizing PDL1. Gastroenterology 2019, 156, 1849–1861. [Google Scholar] [CrossRef] [PubMed]
  104. Choi, H.; Deng, J.; Li, S.; Silk, T.; Dong, L.; Brea, E.J.; Houghton, S.; Redmond, D.; Zhong, H.; Boiarsky, J.; et al. Pulsatile MEK Inhibition Improves Anti-tumor Immunity and T Cell Function in Murine Kras Mutant Lung Cancer. Cell Rep. 2019, 27, 806–819.e5. [Google Scholar] [CrossRef]
  105. Yang, H.; Bueso-Ramos, C.; DiNardo, C.; Estecio, M.R.; Davanlou, M.; Geng, Q.R.; Fang, Z.; Nguyen, M.; Pierce, S.; Wei, Y.; et al. Expression of PD-L1, PD-L2, PD-1 and CTLA4 in myelodysplastic syndromes is enhanced by treatment with hypomethylating agents. Leukemia 2014, 28, 1280–1288. [Google Scholar] [CrossRef]
  106. Wang, L.; Amoozgar, Z.; Huang, J.; Saleh, M.H.; Xing, D.; Orsulic, S.; Goldberg, M.S. Decitabine Enhances Lymphocyte Migration and Function and Synergizes with CTLA-4 Blockade in a Murine Ovarian Cancer Model. Cancer Immunol. Res. 2015, 3, 1030–1041. [Google Scholar] [CrossRef]
  107. Zhang, M.; Zhang, L.; Hei, R.; Li, X.; Cai, H.; Wu, X.; Zheng, Q.; Cai, C. CDK inhibitors in cancer therapy, an overview of recent development. Am. J. Cancer Res. 2021, 11, 1913–1935. [Google Scholar]
  108. Bonelli, P.; Tuccillo, F.M.; Borrelli, A.; Schiattarella, A.; Buonaguro, F.M. CDK/CCN and CDKI Alterations for Cancer Prognosis and Therapeutic Predictivity. BioMed Res. Int. 2014, 2014, e361020. [Google Scholar] [CrossRef]
  109. Shan, W.; Yuan, J.; Hu, Z.; Jiang, J.; Wang, Y.; Loo, N.; Fan, L.; Tang, Z.; Zhang, T.; Xu, M.; et al. Systematic Characterization of Recurrent Genomic Alterations in Cyclin-Dependent Kinases Reveals Potential Therapeutic Strategies for Cancer Treatment. Cell Rep. 2020, 32, 107884. [Google Scholar] [CrossRef]
  110. Yuan, Y.; Lee, J.S.; Yost, S.E.; Frankel, P.H.; Ruel, C.; Egelston, C.A.; Guo, W.; Padam, S.; Tang, A.; Martinez, N.; et al. Phase I/II trial of palbociclib, pembrolizumab and letrozole in patients with hormone receptor-positive metastatic breast cancer. Eur. J. Cancer 2021, 154, 11–20. [Google Scholar] [CrossRef]
  111. Tolaney, S.M.; Kabos, P.; Dickler, M.N.; Gianni, L.; Jansen, V.; Lu, Y.; Young, S.; Rugo, H.S. Updated efficacy, safety, & PD-L1 status of patients with HR+, HER2- metastatic breast cancer administered abemaciclib plus pembrolizumab. J. Clin. Oncol. 2018, 36, 1059. [Google Scholar] [CrossRef] [Green Version]
  112. Pujol, J.-L.; Vansteenkiste, J.; Paz-Ares Rodríguez, L.; Gregorc, V.; Mazieres, J.; Awad, M.; Jänne, P.A.; Chisamore, M.; Hossain, A.M.; Chen, Y.; et al. Abemaciclib in Combination With Pembrolizumab for Stage IV KRAS-Mutant or Squamous NSCLC: A Phase 1b Study. JTO Clin. Res. Rep. 2021, 2, 100234. [Google Scholar] [CrossRef] [PubMed]
  113. Liu, P.; Wang, Y.; Li, X. Targeting the untargetable KRAS in cancer therapy. Acta Pharm. Sinica B 2019, 9, 871–879. [Google Scholar] [CrossRef] [PubMed]
  114. Hauschild, A.; Grob, J.-J.; Demidov, L.V.; Jouary, T.; Gutzmer, R.; Millward, M.; Rutkowski, P.; Blank, C.U.; Miller, W.H.; Kaempgen, E.; et al. Dabrafenib in BRAF-mutated metastatic melanoma: A multicentre, open-label, phase 3 randomised controlled trial. Lancet 2012, 380, 358–365. [Google Scholar] [CrossRef]
  115. Hyman, D.M.; Puzanov, I.; Subbiah, V.; Faris, J.E.; Chau, I.; Blay, J.-Y.; Wolf, J.; Raje, N.S.; Diamond, E.L.; Hollebecque, A.; et al. Vemurafenib in Multiple Nonmelanoma Cancers with BRAF V600 Mutations. N. Engl. J. Med. 2015, 373, 726–736. [Google Scholar] [CrossRef]
  116. Planchard, D.; Kim, T.M.; Mazieres, J.; Quoix, E.; Riely, G.; Barlesi, F.; Souquet, P.-J.; Smit, E.F.; Groen, H.J.M.; Kelly, R.J.; et al. Dabrafenib in patients with BRAF(V600E)-positive advanced non-small-cell lung cancer: A single-arm, multicentre, open-label, phase 2 trial. Lancet Oncol. 2016, 17, 642–650. [Google Scholar] [CrossRef]
  117. Robert, C.; Flaherty, K.T.; Hersey, P.; Nathan, P.D.; Garbe, C.; Milhem, M.M.; Demidov, L.V.; Hassel, J.C.; Rutkowski, P.; Mohr, P.; et al. METRIC phase III study: Efficacy of trametinib (T), a potent and selective MEK inhibitor (MEKi), in progression-free survival (PFS) and overall survival (OS), compared with chemotherapy (C) in patients (pts) with BRAFV600E/K mutant advanced or metastatic melanoma (MM). J. Clin. Oncol. 2012, 30, LBA8509. [Google Scholar] [CrossRef]
  118. Long, G.V.; Stroyakovskiy, D.; Gogas, H.; Levchenko, E.; de Braud, F.; Larkin, J.; Garbe, C.; Jouary, T.; Hauschild, A.; Grob, J.-J.; et al. Dabrafenib and trametinib versus dabrafenib and placebo for Val600 BRAF-mutant melanoma: A multicentre, double-blind, phase 3 randomised controlled trial. Lancet 2015, 386, 444–451. [Google Scholar] [CrossRef]
  119. Planchard, D.; Smit, E.F.; Groen, H.J.M.; Mazieres, J.; Besse, B.; Helland, Å.; Giannone, V.; D’Amelio, A.M.; Zhang, P.; Mookerjee, B.; et al. Dabrafenib plus trametinib in patients with previously untreated BRAFV600E-mutant metastatic non-small-cell lung cancer: An open-label, phase 2 trial. Lancet Oncol. 2017, 18, 1307–1316. [Google Scholar] [CrossRef]
  120. Zhang, Z.; Richmond, A.; Yan, C. Immunomodulatory Properties of PI3K/AKT/mTOR and MAPK/MEK/ERK Inhibition Augment Response to Immune Checkpoint Blockade in Melanoma and Triple-Negative Breast Cancer. Int. J. Mol. Sci. 2022, 23, 7353. [Google Scholar] [CrossRef]
  121. Candido, S.; Salemi, R.; Piccinin, S.; Falzone, L.; Libra, M. The PIK3CA H1047R Mutation Confers Resistance to BRAF and MEK Inhibitors in A375 Melanoma Cells through the Cross-Activation of MAPK and PI3K–Akt Pathways. Pharmaceutics 2022, 14, 590. [Google Scholar] [CrossRef] [PubMed]
  122. Atkins, M.B.; Lee, S.J.; Chmielowski, B.; Ribas, A.; Tarhini, A.A.; Truong, T.-G.; Davar, D.; O’Rourke, M.A.; Curti, B.D.; Brell, J.M.; et al. DREAMseq (Doublet, Randomized Evaluation in Advanced Melanoma Sequencing): A phase III trial—ECOG-ACRIN EA6134. J. Clin. Oncol. 2021, 39, 356154. [Google Scholar] [CrossRef]
  123. Ascierto, P.A.; Ferrucci, P.F.; Fisher, R.; Del Vecchio, M.; Atkinson, V.; Schmidt, H.; Schachter, J.; Queirolo, P.; Long, G.V.; Di Giacomo, A.M.; et al. Dabrafenib, trametinib and pembrolizumab or placebo in BRAF-mutant melanoma. Nat. Med. 2019, 25, 941–946. [Google Scholar] [CrossRef]
  124. Ascierto, P.A.; McArthur, G.A.; Dréno, B.; Atkinson, V.; Liszkay, G.; Di Giacomo, A.M.; Mandalà, M.; Demidov, L.; Stroyakovskiy, D.; Thomas, L.; et al. Cobimetinib combined with vemurafenib in advanced BRAFV600-mutant melanoma (coBRIM): Updated efficacy results from a randomised, double-blind, phase 3 trial. Lancet Oncol. 2016, 17, 1248–1260. [Google Scholar] [CrossRef]
  125. Gogas, H.; Dréno, B.; Larkin, J.; Demidov, L.; Stroyakovskiy, D.; Eroglu, Z.; Ferrucci, P.F.; Pigozzo, J.; Rutkowski, P.; Mackiewicz, J.; et al. Cobimetinib plus atezolizumab in BRAFV600 wild-type melanoma: Primary results from the randomized phase III IMspire170 study. Ann. Oncol. 2021, 32, 384–394. [Google Scholar] [CrossRef] [PubMed]
  126. Gutzmer, R.; Stroyakovskiy, D.; Gogas, H.; Robert, C.; Lewis, K.; Protsenko, S.; Pereira, R.P.; Eigentler, T.; Rutkowski, P.; Demidov, L.; et al. Atezolizumab, vemurafenib, and cobimetinib as first-line treatment for unresectable advanced BRAFV600 mutation-positive melanoma (IMspire150): Primary analysis of the randomised, double-blind, placebo-controlled, phase 3 trial. Lancet 2020, 395, 1835–1844. [Google Scholar] [CrossRef]
  127. Yarchoan, M.; Cope, L.; Ruggieri, A.N.; Anders, R.A.; Noonan, A.M.; Goff, L.W.; Goyal, L.; Lacy, J.; Li, D.; Patel, A.K.; et al. Multicenter randomized phase II trial of atezolizumab with or without cobimetinib in biliary tract cancers. J. Clin. Investig. 2021, 131, e152670. [Google Scholar] [CrossRef]
  128. Liu, S.V.; Hall, R.D.; Saltos, A.N.; Otterson, G.A.; Tan, M.T.; Gnjatic, S.; Gentzler, R.D.; Tanvetyanon, T.; Chen, H.X.; Sharon, E. A phase II study of atezolizumab and cobimetinib in PD-1/PD-L1 inhibitor resistant or refractory non-small cell lung cancer: ETCTN #10166. J. Clin. Oncol. 2020, 38, TPS9638. [Google Scholar] [CrossRef]
  129. Subramaniam, D.; He, A.R.; Hwang, J.; Deeken, J.; Pishvaian, M.; Hartley, M.L.; Marshall, J.L. Irreversible multitargeted ErbB family inhibitors for therapy of lung and breast cancer. Curr. Cancer Drug Targets 2015, 14, 775–793. [Google Scholar] [CrossRef]
  130. Roskoski, R. Small molecule inhibitors targeting the EGFR/ErbB family of protein-tyrosine kinases in human cancers. Pharmacol. Res. 2019, 139, 395–411. [Google Scholar] [CrossRef]
  131. Creelan, B.C.; Yeh, T.C.; Kim, S.-W.; Nogami, N.; Kim, D.-W.; Chow, L.Q.M.; Kanda, S.; Taylor, R.; Tang, W.; Tang, M.; et al. A Phase 1 study of gefitinib combined with durvalumab in EGFR TKI-naive patients with EGFR mutation-positive locally advanced/metastatic non-small-cell lung cancer. Br. J. Cancer 2021, 124, 383–390. [Google Scholar] [CrossRef] [PubMed]
  132. Riudavets, M.; Naigeon, M.; Texier, M.; Dorta, M.; Barlesi, F.; Mazieres, J.; Varga, A.; Cassard, L.; Boselli, L.; Grivel, J.; et al. Gefitinib plus tremelimumab combination in refractory non-small cell lung cancer patients harbouring EGFR mutations: The GEFTREM phase I trial. Lung Cancer 2022, 166, 255–264. [Google Scholar] [CrossRef] [PubMed]
  133. Yang, J.C.-H.; Gadgeel, S.M.; Sequist, L.V.; Wu, C.-L.; Papadimitrakopoulou, V.A.; Su, W.-C.; Fiore, J.; Saraf, S.; Raftopoulos, H.; Patnaik, A. Pembrolizumab in Combination With Erlotinib or Gefitinib as First-Line Therapy for Advanced NSCLC With Sensitizing EGFR Mutation. J. Thorac. Oncol. 2019, 14, 553–559. [Google Scholar] [CrossRef] [PubMed]
  134. Yamaguchi, O.; Kaira, K.; Kawasaki, T.; Mouri, A.; Hashimoto, K.; Shiono, A.; Shinomiya, S.; Miura, Y.; Nishihara, F.; Murayama, Y.; et al. Severe hepatotoxicity due to osimertinib after nivolumab therapy in patients with non-small cell lung cancer harboring EGFR mutation. Thorac. Cancer 2020, 11, 1045–1051. [Google Scholar] [CrossRef]
  135. Sacco, A.G.; Chen, R.; Worden, F.P.; Wong, D.J.L.; Adkins, D.; Swiecicki, P.; Chai-Ho, W.; Oppelt, P.; Ghosh, D.; Bykowski, J.; et al. Pembrolizumab plus cetuximab in patients with recurrent or metastatic head and neck squamous cell carcinoma: An open-label, multi-arm, non-randomised, multicentre, phase 2 trial. Lancet Oncol. 2021, 22, 883–892. [Google Scholar] [CrossRef]
  136. Boland, P.M.; Hutson, A.; Maguire, O.; Minderman, H.; Fountzilas, C.; Iyer, R.V. A phase Ib/II study of cetuximab and pembrolizumab in RAS-wt mCRC. J. Clin. Oncol. 2018, 36, 834. [Google Scholar] [CrossRef]
  137. Lee, M.S.; Loehrer, P.J.; Imanirad, I.; Cohen, S.; Ciombor, K.K.; Moore, D.T.; Carlson, C.A.; Sanoff, H.K.; McRee, A.J. Phase II study of ipilimumab, nivolumab, and panitumumab in patients with KRAS/NRAS/BRAF wild-type (WT) microsatellite stable (MSS) metastatic colorectal cancer (mCRC). J. Clin. Oncol. 2021, 39, 7. [Google Scholar] [CrossRef]
  138. Besse, B.; Garrido, P.; Cortot, A.B.; Johnson, M.; Murakami, H.; Gazzah, A.; Gil, M.; Bennouna, J. Efficacy and safety of necitumumab and pembrolizumab combination therapy in patients with Stage IV non-small cell lung cancer. Lung Cancer 2020, 142, 63–69. [Google Scholar] [CrossRef]
  139. Krishnamurti, U.; Silverman, J.F. HER2 in breast cancer: A review and update. Adv. Anat. Pathol. 2014, 21, 100–107. [Google Scholar] [CrossRef]
  140. Kao, H.-F.; Liao, B.-C.; Huang, Y.-L.; Huang, H.-C.; Chen, C.-N.; Chen, T.-C.; Hong, Y.-J.; Chan, C.-Y.; Chia, J.-S.; Hong, R.-L. Afatinib and Pembrolizumab for Recurrent or Metastatic Head and Neck Squamous Cell Carcinoma (ALPHA Study): A Phase II Study with Biomarker Analysis. Clin. Cancer Res. 2022, 28, 1560–1571. [Google Scholar] [CrossRef]
  141. Loi, S.; Giobbie-Hurder, A.; Gombos, A.; Bachelot, T.; Hui, R.; Curigliano, G.; Campone, M.; Biganzoli, L.; Bonnefoi, H.; Jerusalem, G.; et al. Pembrolizumab plus trastuzumab in trastuzumab-resistant, advanced, HER2-positive breast cancer (PANACEA): A single-arm, multicentre, phase 1b–2 trial. Lancet Oncol. 2019, 20, 371–382. [Google Scholar] [CrossRef]
  142. Janjigian, Y.Y.; Maron, S.B.; Chatila, W.K.; Millang, B.; Chavan, S.S.; Alterman, C.; Chou, J.F.; Segal, M.F.; Simmons, M.Z.; Momtaz, P.; et al. First-line pembrolizumab and trastuzumab in HER2-positive oesophageal, gastric, or gastro-oesophageal junction cancer: An open-label, single-arm, phase 2 trial. Lancet Oncol. 2020, 21, 821–831. [Google Scholar] [CrossRef]
  143. Chen, A. PARP inhibitors: Its role in treatment of cancer. Chin. J. Cancer 2011, 30, 463–471. [Google Scholar] [CrossRef] [PubMed]
  144. 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]
  145. 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. Off. J. Am. Assoc. Cancer Res. 2020, 26, 4268–4279. [Google Scholar] [CrossRef]
  146. 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. ImmunoTherapy Cancer 2018, 6, 141. [Google Scholar] [CrossRef]
  147. Fumet, J.-D.; Limagne, E.; Thibaudin, M.; Truntzer, C.; Bertaut, A.; Rederstorff, E.; Ghiringhelli, F. Precision medicine phase II study evaluating the efficacy of a double immunotherapy by durvalumab and tremelimumab combined with olaparib in patients with solid cancers and carriers of homologous recombination repair genes mutation in response or stable after olaparib treatment. BMC Cancer 2020, 20, 748. [Google Scholar] [CrossRef]
  148. 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]
  149. Taeger, J.; Moser, C.; Hellerbrand, C.; Mycielska, M.E.; Glockzin, G.; Schlitt, H.J.; Geissler, E.K.; Stoeltzing, O.; Lang, S.A. Targeting FGFR/PDGFR/VEGFR impairs tumor growth, angiogenesis, and metastasis by effects on tumor cells, endothelial cells, and pericytes in pancreatic cancer. Mol. Cancer Ther. 2011, 10, 2157–2167. [Google Scholar] [CrossRef]
  150. Chu, T.; Zhong, R.; Zhong, H.; Zhang, B.; Zhang, W.; Shi, C.; Qian, J.; Zhang, Y.; Chang, Q.; Zhang, X.; et al. Phase 1b Study of Sintilimab Plus Anlotinib as First-line Therapy in Patients With Advanced NSCLC. J. Thorac. Oncol. 2021, 16, 643–652. [Google Scholar] [CrossRef]
  151. Liu, J.; Jiang, Z.; Li, Q.; Li, Y.; Liu, Q.; Song, E. Efficacy and safety of anti-PD-1 antibody SHR-1210 combined with apatinib in patients with advanced triple-negative breast cancer. J. Clin. Oncol. 2019, 37, 1066. [Google Scholar] [CrossRef]
  152. Xu, J.; Zhang, Y.; Jia, R.; Yue, C.; Chang, L.; Liu, R.; Zhang, G.; Zhao, C.; Zhang, Y.; Chen, C.; et al. Anti-PD-1 Antibody SHR-1210 Combined with Apatinib for Advanced Hepatocellular Carcinoma, Gastric, or Esophagogastric Junction Cancer: An Open-label, Dose Escalation and Expansion Study. Clin. Cancer Res. Off. J. Am. Assoc. Cancer Res. 2019, 25, 515–523. [Google Scholar] [CrossRef] [PubMed]
  153. Ren, C.; Mai, Z.-J.; Jin, Y.; He, M.-M.; Wang, Z.-Q.; Luo, H.-Y.; Zhang, D.-S.; Wu, C.-Y.; Wang, F.; Xu, R.-H. Anti-PD-1 antibody SHR-1210 plus apatinib for metastatic colorectal cancer: A prospective, single-arm, open-label, phase II trial. Am. J. Cancer Res. 2020, 10, 2946–2954. [Google Scholar] [PubMed]
  154. Powles, T.; Plimack, E.R.; Soulières, D.; Waddell, T.; Stus, V.; Gafanov, R.; Nosov, D.; Pouliot, F.; Melichar, B.; Vynnychenko, I.; et al. Pembrolizumab plus axitinib versus sunitinib monotherapy as first-line treatment of advanced renal cell carcinoma (KEYNOTE-426): Extended follow-up from a randomised, open-label, phase 3 trial. Lancet Oncol. 2020, 21, 1563–1573. [Google Scholar] [CrossRef]
  155. Choueiri, T.K.; Powles, T.; Burotto, M.; Escudier, B.; Bourlon, M.T.; Zurawski, B.; Oyervides Juárez, V.M.; Hsieh, J.J.; Basso, U.; Shah, A.Y.; et al. Nivolumab plus Cabozantinib versus Sunitinib for Advanced Renal-Cell Carcinoma. N. Engl. J. Med. 2021, 384, 829–841. [Google Scholar] [CrossRef]
  156. Finn, R.S.; Ikeda, M.; Zhu, A.X.; Sung, M.W.; Baron, A.D.; Kudo, M.; Okusaka, T.; Kobayashi, M.; Kumada, H.; Kaneko, S.; et al. Phase Ib Study of Lenvatinib Plus Pembrolizumab in Patients With Unresectable Hepatocellular Carcinoma. J. Clin. Oncol. 2020, 38, 2960–2970. [Google Scholar] [CrossRef]
  157. Haugen, B.; French, J.; Worden, F.P.; Konda, B.; Sherman, E.J.; Dadu, R.; Gianoukakis, A.G.; Wolfe, E.G.; Foster, N.R.; Bowles, D.W.; et al. Lenvatinib plus pembrolizumab combination therapy in patients with radioiodine-refractory (RAIR), progressive differentiated thyroid cancer (DTC): Results of a multicenter phase II international thyroid oncology group trial. J. Clin. Oncol. 2020, 38, 6512. [Google Scholar] [CrossRef]
  158. Kawazoe, A.; Fukuoka, S.; Nakamura, Y.; Kuboki, Y.; Wakabayashi, M.; Nomura, S.; Mikamoto, Y.; Shima, H.; Fujishiro, N.; Higuchi, T.; et al. Lenvatinib plus pembrolizumab in patients with advanced gastric cancer in the first-line or second-line setting (EPOC1706): An open-label, single-arm, phase 2 trial. Lancet Oncol. 2020, 21, 1057–1065. [Google Scholar] [CrossRef]
  159. Lin, J.; Yang, X.; Long, J.; Zhao, S.; Mao, J.; Wang, D.; Bai, Y.; Bian, J.; Zhang, L.; Yang, X.; et al. Pembrolizumab combined with lenvatinib as non-first-line therapy in patients with refractory biliary tract carcinoma. Hepatobiliary Surg. Nutr. 2020, 9, 414–424. [Google Scholar] [CrossRef]
  160. Llovet, J.; Shepard, K.V.; Finn, R.S.; Ikeda, M.; Sung, M.; Baron, A.D.; Kudo, M.; Okusaka, T.; Kobayashi, M.; Kumada, H.; et al. 747P—A phase Ib trial of lenvatinib (LEN) plus pembrolizumab (PEMBRO) in unresectable hepatocellular carcinoma (uHCC): Updated results. Ann. Oncol. 2019, 30, v286–v287. [Google Scholar] [CrossRef]
  161. Cousin, S.; Cantarel, C.; Guegan, J.-P.; Gomez-Roca, C.; Metges, J.-P.; Adenis, A.; Pernot, S.; Bellera, C.; Kind, M.; Auzanneau, C.; et al. Regorafenib-Avelumab Combination in Patients with Microsatellite Stable Colorectal Cancer (REGOMUNE): A Single-arm, Open-label, Phase II Trial. Clin. Cancer Res. Off. J. Am. Assoc. Cancer Res. 2021, 27, 2139–2147. [Google Scholar] [CrossRef] [PubMed]
  162. Huang, J.; Guo, Y.; Huang, W.; Hong, X.; Quan, Y.; Lin, L.; Zhou, J.; Liang, L.; Zhang, Y.; Zhou, J.; et al. Regorafenib Combined with PD-1 Blockade Immunotherapy versus Regorafenib as Second-Line Treatment for Advanced Hepatocellular Carcinoma: A Multicenter Retrospective Study. J. Hepatocell. Carcinoma 2022, 9, 157–170. [Google Scholar] [CrossRef] [PubMed]
  163. Gutierrez, M.; Subbiah, V.; Nemunaitis, J.J.; Mettu, N.B.; Papadopoulos, K.P.; Barve, M.A.; Féliz, L.; Lihou, C.F.; Tian, C.; Ji, T.; et al. Safety and efficacy of pemigatinib plus pembrolizumab combination therapy in patients (pts) with advanced malignancies: Results from FIGHT-101, an open-label phase I/II study. J. Clin. Oncol. 2020, 38, 3606. [Google Scholar] [CrossRef]
  164. Zhu, X.D.; Huang, C.; Shen, Y.H.; Ji, Y.; Ge, N.L.; Qu, X.D.; Chen, L.; Shi, W.K.; Li, M.L.; Zhu, J.J.; et al. Downstaging and Resection of Initially Unresectable Hepatocellular Carcinoma with Tyrosine Kinase Inhibitor and Anti-PD-1 Antibody Combinations. Liver Cancer 2021, 10, 320–329. [Google Scholar] [CrossRef] [PubMed]
  165. Finn, R.S.; Qin, S.; Ikeda, M.; Galle, P.R.; Ducreux, M.; Kim, T.-Y.; Kudo, M.; Breder, V.; Merle, P.; Kaseb, A.O.; et al. Atezolizumab plus Bevacizumab in Unresectable Hepatocellular Carcinoma. N. Engl. J. Med. 2020, 382, 1894–1905. [Google Scholar] [CrossRef] [PubMed]
  166. Motzer, R.J.; Powles, T.; Atkins, M.B.; Escudier, B.; McDermott, D.F.; Suarez, C.; Bracarda, S.; Stadler, W.M.; Donskov, F.; Lee, J.-L.; et al. IMmotion151: A Randomized Phase III Study of Atezolizumab Plus Bevacizumab vs Sunitinib in Untreated Metastatic Renal Cell Carcinoma (mRCC). J. Clin. Oncol. 2018, 36, 578. [Google Scholar] [CrossRef]
  167. Hellmann, M.D.; Jänne, P.A.; Opyrchal, M.; Hafez, N.; Raez, L.E.; Gabrilovich, D.I.; Wang, F.; Trepel, J.B.; Lee, M.-J.; Yuno, A.; et al. Entinostat plus Pembrolizumab in Patients with Metastatic NSCLC Previously Treated with Anti–PD-(L)1 Therapy. Clin. Cancer Res. 2021, 27, 1019–1028. [Google Scholar] [CrossRef]
  168. Ny, L.; Jespersen, H.; Karlsson, J.; Alsén, S.; Filges, S.; All-Eriksson, C.; Andersson, B.; Carneiro, A.; Helgadottir, H.; Levin, M.; et al. The PEMDAC phase 2 study of pembrolizumab and entinostat in patients with metastatic uveal melanoma. Nat. Commun. 2021, 12, 5155. [Google Scholar] [CrossRef]
  169. Wang, C.; Liu, Y.; Dong, L.; Li, X.; Yang, Q.; Brock, M.V.; Mei, Q.; Liu, J.; Chen, M.; Shi, F.; et al. Efficacy of Decitabine plus Anti-PD-1 Camrelizumab in Patients with Hodgkin Lymphoma Who Progressed or Relapsed after PD-1 Blockade Monotherapy. Clin. Cancer Res. 2021, 27, 2782–2791. [Google Scholar] [CrossRef]
  170. Saxena, K.; Herbrich, S.M.; Pemmaraju, N.; Kadia, T.M.; DiNardo, C.D.; Borthakur, G.; Pierce, S.A.; Jabbour, E.; Wang, S.A.; Bueso-Ramos, C.; et al. A phase 1b/2 study of azacitidine with PD-L1 antibody avelumab in relapsed/refractory acute myeloid leukemia. Cancer 2021, 127, 3761–3771. [Google Scholar] [CrossRef]
  171. Petroni, G.; Buqué, A.; Coussens, L.M.; Galluzzi, L. Targeting oncogene and non-oncogene addiction to inflame the tumour microenvironment. Nat. Rev. Drug Discov. 2022, 21, 440–462. [Google Scholar] [CrossRef]
  172. Palmer, A.C.; Sorger, P.K. Combination Cancer Therapy Can Confer Benefit via Patient-to-Patient Variability without Drug Additivity or Synergy. Cell 2017, 171, 1678–1691.e1613. [Google Scholar] [CrossRef] [PubMed]
  173. Van Herck, Y.; Feyaerts, A.; Alibhai, S.; Papamichael, D.; Decoster, L.; Lambrechts, Y.; Pinchuk, M.; Bechter, O.; Herrera-Caceres, J.; Bibeau, F.; et al. Is cancer biology different in older patients? Lancet Healthy Longev. 2021, 2, e663–e677. [Google Scholar] [CrossRef]
  174. Fane, M.; Weeraratna, A.T. How the ageing microenvironment influences tumour progression. Nat. Rev. Cancer 2020, 20, 89–106. [Google Scholar] [CrossRef] [PubMed]
  175. Howie, L.J.; Singh, H.; Bloomquist, E.; Wedam, S.; Amiri-Kordestani, L.; Tang, S.; Sridhara, R.; Sanchez, J.; Prowell, T.M.; Kluetz, P.G.; et al. Outcomes of Older Women With Hormone Receptor–Positive, Human Epidermal Growth Factor Receptor–Negative Metastatic Breast Cancer Treated With a CDK4/6 Inhibitor and an Aromatase Inhibitor: An FDA Pooled Analysis. J. Clin. Oncol. 2019, 37, 3475–3483. [Google Scholar] [CrossRef] [PubMed]
Figure 1. Modulatory Effects of Targeted Therapy on Immune Cells. The molecularly targeted agents function as immunostimulatory (the green block diagram) or immunosuppressive (the red block diagram) modulators in TIME. The color of each little arrow matches the corresponding drug category in the middle of figure. ADCC, antibody-dependent cell cytotoxicity; ADCP, antibody-dependent cellular phagocytosis; AKT, V-akt murine thymoma viral oncogene homolog; BTK, Bruton’s tyrosine kinase; CDK, cycle-dependent kinase; EGFR, epidermal growth factor receptor; FGFR, fibroblast growth factor receptor; HER, human epidermal growth factor receptor; KDR, kinase insert domain receptor; MAPK, mitogen-activated protein kinases; MDM2, mouse double minute 2; MDSC, myeloid-derived suppressor cell; mTOR, mechanistic target of rapamycin; PARP, poly(ADP-ribose) polymerase; PDGFR, platelet-derived growth factors receptor; PI3K, phosphatidylinositol-3-kinase; TAM, tumor-associated macrophage; TIME, tumor immune microenvironment; TGF-β, transforming growth factor-β; Treg, regulatory T cell; VEGFR, vascular endothelial growth factor receptor (created with BioRender.com (accessed on 11 August 2022)).
Figure 1. Modulatory Effects of Targeted Therapy on Immune Cells. The molecularly targeted agents function as immunostimulatory (the green block diagram) or immunosuppressive (the red block diagram) modulators in TIME. The color of each little arrow matches the corresponding drug category in the middle of figure. ADCC, antibody-dependent cell cytotoxicity; ADCP, antibody-dependent cellular phagocytosis; AKT, V-akt murine thymoma viral oncogene homolog; BTK, Bruton’s tyrosine kinase; CDK, cycle-dependent kinase; EGFR, epidermal growth factor receptor; FGFR, fibroblast growth factor receptor; HER, human epidermal growth factor receptor; KDR, kinase insert domain receptor; MAPK, mitogen-activated protein kinases; MDM2, mouse double minute 2; MDSC, myeloid-derived suppressor cell; mTOR, mechanistic target of rapamycin; PARP, poly(ADP-ribose) polymerase; PDGFR, platelet-derived growth factors receptor; PI3K, phosphatidylinositol-3-kinase; TAM, tumor-associated macrophage; TIME, tumor immune microenvironment; TGF-β, transforming growth factor-β; Treg, regulatory T cell; VEGFR, vascular endothelial growth factor receptor (created with BioRender.com (accessed on 11 August 2022)).
Pharmaceutics 14 01768 g001
Table 1. Clinical trials of ICIs plus targeted agents targeting BRAF/MEK, CDK, or ErbB.
Table 1. Clinical trials of ICIs plus targeted agents targeting BRAF/MEK, CDK, or ErbB.
Cancer TypeTargeted AgentsCombination AgentsPhaseControlPrimary EndpointNCT Number
BRAF/MEK Inhibitors
Melanoma *Cobimetinib + VemurafenibAtezolizumabIIICobimetinib + Vemurafenib + PFS (vs. control): 15.1 vs. 10.6 months, HR = 0.78NCT02908672
placebo
MelanomaTrametinib + Dabrafenib Ipilimumab + NivolumabIIIArm A: N/I + D/T2-year OS rate (Arm A vs. Arm B): 72% vs. 52%NCT02224781
Arm B: D/T + N/I
CDK Inhibitors
Breast CancerPalbociclibPembrolizumab + LetrozoleI/II-ORR: 56%NCT02778685
EGFR Inhibitors
HNSCCCetuximabPembrolizumabII-ORR: 45% (95% CI 28–62)NCT03082534
CRCPanitumumabIpilimumab + NivolumabII-ORR: 35% (95% CI 21–48)NCT03442569
CRCCetuximabPembrolizumabIb/II-A manageable safety profileNCT02713373
HER2
HNSCCAfatinibPembrolizumabII-High PD-L1 expression (TPS ≥ 50 ORR: 71%, CPS ≥ 20 ORR: 63%);NCT03695510
EGFR amplification (ORR: 100%);
MTAP loss or mutation (ORR: 0%)
Oesophageal, G/GEJ CancerTrastuzumabPembrolizumabII-6-months PFSR: 70% (95% CI 54–83)NCT02954536
Breast CancerTrastuzumabPembrolizumabIb/II-ORR: 15% (90% CI 7–29)NCT02129556
*: Approved by FDA. Source: http://www.clinicaltrials.gov (accessed on 29 July 2022). CRC, colorectal cancer; CDK, cycle-dependent kinase; CI, confidence interval; CPS, combined positive score; EGFR, epidermal growth factor receptor; HNSCC, head and neck squamous cell carcinoma; HR, hazard ratio; MEK, mitogen-activated protein kinase; MTAP, methylthioadenosine phosphorylase; ORR, overall response rate; PFS, progression-free survival; PFSR, progression-free survival rate; TPS, tumor proportion score.
Table 2. Clinical trials of ICIs plus targeted agents targeting FGFR/PDGFR/VEGFR.
Table 2. Clinical trials of ICIs plus targeted agents targeting FGFR/PDGFR/VEGFR.
Cancer TypeTargeted AgentsCombination AgentsPhaseControl Primary EndpointNCT Number
Endometrial Cancer *LenvatinibPembrolizumabIIIChemotherapyMedian OS (vs. control): 17.4 vs. 12.0 months, HR = 0.68NCT03517449
Median PFS (vs. control): 6.6 vs. 3.8 months, HR = 0.60
HCC *BevacizumabAtezolizumabIIISorafenibMedian OS (vs. control): 19.2 vs. 13.4 months, HR = 0.66NCT03434379
Median PFS (vs. control): 6.9 vs. 4.3 months, HR = 0.65
RCC *LenvatinibPembrolizumabIIILenvatinib + PFS (vs. control): 23.9 vs. 9.2 months, HR = 0.39NCT02811861
Sunitinib
RCC *AxitinibPembrolizumabIIISunitinibOS (vs. control): median not reached vs. 35·7 months, HR = 0.68 NCT02853331
RCC *CabozantinibNivolumabIIISunitinibPFS (vs. control): 16.6 vs. 8.3 months, HR = 0.51NCT03141177
RCCBevacizumabAtezolizumabIIISunitinibPD-L1 positive group median PFS (vs. control): 11.2 vs. 7.7 months, HR = 0.74NCT02420821
Breast CancerApatinibCamrelizumabII-ORR: 43.3% (95% CI 25.5–62.6)NCT03394287
CRCRegorafenibAvelumabII-CR, PR: 0; SD: 57.5%NCT03475953
DTCLenvatinibPembrolizumabII-ORR: 62%NCT02973997
Endometrial CancerLenvatinibPembrolizumabII-ORR: 38% (95% CI 28.8–47.8)NCT02501096
GCLenvatinibPembrolizumabII-ORR: 69% (95% CI 49–85)NCT03609359
Ovarian CancerBevacizumabNivolumabII-ORR: 21%NCT02873962
CRCRegorafenibToripalimabIb/II-ORR: 15.2% (95% CI 5.7–32.7)NCT03946917
PancancerPemigatinibPembrolizumabI/II-A manageable safety profile and pharmacodynamic and clinical activityNCT02393248
SarcomasSunitinibNivolumabIb/II-6-months PFSR: 48% (95% CI 41–55)NCT03277924
Gastric Cancer/CRCRegorafenibNivolumabIb-Safety: Regorafenib 80 mg plus nivolumabNCT03406871
HCCLenvatinibPembrolizumabIb-Promising antitumor activity with a tolerable safety profileNCT03006926
NSCLCAnlotinibSintilimabIb-ORR: 72.7% (95% CI 49.8–89.3)NCT03628521
HCCApatinibCamrelizumabI-ORR: 30.8% (95% CI 17–47.6)NCT02942329
NSCLC, G/GEJ Cancer, UCRamucirumabPembrolizumabI-A manageable safety profile with favorable antitumor activityNCT02443324
*: Approved by FDA. Source: http://www.clinicaltrials.gov (accessed on 29 July 2022). CI, confidence interval; CR, complete remission; CRC, colorectal cancer; DTC, differentiated thyroid cancer; GC, gastric cancer; HCC, hepatocellular carcinoma; HR, hazard ratio; NSCLC, non-small cell lung cancer; ORR, overall response rate; OS, overall survival; PFS, progression-free survival; PFSR, progression-free survival rate; PR, partial remission; RCC, renal cell carcinoma; UC, urothelial carcinoma.
Table 3. Clinical trials of ICIs plus epigenetic agents.
Table 3. Clinical trials of ICIs plus epigenetic agents.
Cancer TypeTargeted AgentsCombination AgentsPhaseControlPrimary EndpointNCT Number
Uveal MelanomaEntinostatPembrolizumabII-ORR: 14% (95% CI 3.9–31.7)NCT02697630
AMLAzacitidineNivolumabII-ORR: 33%NCT02397720
Hodgkin LymphomaDecitabineCamrelizumabII-ORR: 60% (95% CI 45–74)NCT02961101
Source: http://www.clinicaltrials.gov (accessed on 29 July 2022). AML, acute myeloid leukemia; CI, confidence interval; ORR, objective response rate.
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Share and Cite

MDPI and ACS Style

Zhang, T.; Zhang, C.; Fu, Z.; Gao, Q. Immune Modulatory Effects of Molecularly Targeted Therapy and Its Repurposed Usage in Cancer Immunotherapy. Pharmaceutics 2022, 14, 1768. https://doi.org/10.3390/pharmaceutics14091768

AMA Style

Zhang T, Zhang C, Fu Z, Gao Q. Immune Modulatory Effects of Molecularly Targeted Therapy and Its Repurposed Usage in Cancer Immunotherapy. Pharmaceutics. 2022; 14(9):1768. https://doi.org/10.3390/pharmaceutics14091768

Chicago/Turabian Style

Zhang, Tiancheng, Chenhao Zhang, Zile Fu, and Qiang Gao. 2022. "Immune Modulatory Effects of Molecularly Targeted Therapy and Its Repurposed Usage in Cancer Immunotherapy" Pharmaceutics 14, no. 9: 1768. https://doi.org/10.3390/pharmaceutics14091768

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