• All
  • Article Title
  • Abstract
  • Keywords
  • Author
  • Institution
  • Open Access
    Review

    Immune-checkpoint inhibitor resistance in patients with cancer

    Luis Cabezón-Gutiérrez 1,2*
    Sara Custodio-Cabello 1,2
    Magda Palka-Kotlowska 1,2
    Beatriz Chacón-Ovejero 3
    Vilma Pacheco-Barcia 1,2

    Explor Immunol. 2025;5:1003196 DOI: https://doi.org/10.37349/ei.2025.1003196

    Received: July 08, 2024 Accepted: April 22, 2025 Published: May 20, 2025

    Academic Editor: Cunte Chen, South China University of Technology, China

    This article belongs to the special issue The Role of Immune Checkpoint Molecules in Cancer and Hematological Malignancies

    Abstract

    The introduction of immune checkpoint inhibitors (ICIs) has transformed the landscape of oncology, offering significant improvements in patient survival and achieving remarkable long-term outcomes. Despite these advances, the therapeutic benefits of ICIs are not universal, and existing biomarkers often fall short in accurately predicting patient responses. A comprehensive understanding of the mechanisms underlying resistance to ICIs is essential for the development of strategies to mitigate these challenges and enhance therapeutic efficacy. This review provides a detailed exploration of the resistance mechanisms associated with ICIs, focusing on the role of the tumor microenvironment and intrinsic tumor cell alterations in mediating both primary and secondary resistance. Furthermore, it evaluates emerging strategies to overcome resistance, including combination therapies and innovative therapeutic approaches. By dissecting the molecular and immunological pathways implicated in ICI resistance, this review aims to highlight novel predictive and prognostic biomarkers and outline optimized therapeutic strategies to maximize the clinical impact of ICIs in cancer management.

    Keywords

    Immune checkpoint inhibitor, immunotherapy resistance, tumor microenvironment, combination therapy

    Introduction

    The emergence of immune checkpoint inhibitors (ICIs) in multiple types of tumors has revolutionized oncology. Monoclonal antibodies targeting the programmed cell death protein 1/programmed death ligand 1 (PD-1/PD-L1) pathway, demonstrate significant clinical efficacy in a range of solid tumors [16]. However, many patients eventually develop resistance to ICIs over time, leading to a loss of efficacy.

    While numerous studies have sought to identify biomarkers predicting primary non-response to ICIs, the mechanisms underlying acquired resistance remain largely unknown.

    Primary resistance mechanisms encompass the absence or loss of tumor antigens [7], alterations in the major histocompatibility complex (MHC) processing pathway [8], low T-cell infiltration, increased expression of vascular endothelial growth factor (VEGF) and immunosuppressive cytokines [9], and mutations in STK11 [10]. PD-L1 expression on tumor cells is regarded as a positive predictive marker [6, 11, 12]; however, many patients fail to respond to ICI treatment despite having tumors with high PD-L1 expression [13].

    Mechanisms contributing to acquired resistance include the upregulation of alternative immune checkpoints, the loss of human leukocyte antigen (HLA) expression, and mutations in β2-microglobulin (β2M) and Janus kinase 1/2 (JAK1/2) [1417]. When patients develop resistance to immunotherapy, there are limited therapeutic options, and a lack of detailed understanding of these resistance mechanisms makes it difficult to guide treatment decisions.

    Currently, data on the mechanisms involved in ICI resistance are lacking. Gaining a deeper understanding of the molecular and immunological processes governing ICIs will aid in identifying new predictive and prognostic biomarkers and establishing optimal therapeutic strategies. In this review, we examine emerging evidence that sheds light on novel mechanisms of both innate and acquired resistance to ICIs and offer insights into potential strategies for overcoming these challenges.

    Cancer immunotherapy: immune checkpoint inhibitors

    The immune system has a key role in the natural history of cancer development, tumor growth and metastasis but tumors have the ability to evade the immune response. Several cell types participate in the interplay of the immune reaction: (1) the “immune synapse” is the physical contact of immune cells by antigen presentation and has helper T cells (the subclasses Th1/Th2 of CD4+ T lymphocytes) and CD8+ T lymphocytes which are able to discriminate non self-antigens vs self-antigens [1820]; (2) natural killer (NK) cells have inhibitory molecules and killer cell immunoglobulin-like receptor (KIR) subtypes [21], NK cells can exert cytotoxicity over with low MHC class 1 expression because they do not need MHC antigen presentation; (3) there are two different phenotypes of macrophages: M1 macrophages that are capable of liberating interferon (IFN) gamma and M2 macrophages and are also responsible for phagocytosis, that can inhibit inflammatory reactions and promote tolerance by the release of cytokines such as transforming growth factor beta (TGF-β), interleukin (IL)-4 and IL-10 [22]; (4) other types of cells that play a role in autoimmunity and cancer are myeloid derived suppressor cells (MDSCs) and FoxP3+ CD25+ CD4+ Treg that dampen cytotoxic T lymphocyte activity [23, 24] and CD4+ T cells that liberate IL-17, the Th17 cells.

    The “immune synapse” refers to the immunological phenomenon based upon the capacity of T lymphocytes to differentiate non-self and self-antigens presented by antigen-presenting cells (APCs) and how the cytotoxic CD8+ T cells are modulated depending on the inhibitory and stimulatory receptors which are, furthermore, regulated by cytokines [20]. The T cell receptor (TCR) complex includes the TCR, the CD4 or CD8 receptor that binds to the MHC and the CD3, macromolecular complex formed by CD3 independent molecules [18], which activates through an intracellular tyrosine-based component that transfers surface signals to intracellular effectors [25]. The TCR needs to adhere to a peptide exhibited by the MHC and there also needs to be a set of costimulatory signals that allow an efficient naive CD8+ T cells activation [20]. Thus, naive CD8+ T cell activation leads to the increase of production of pro-inflammatory cytokines such as IFN gamma and IL-12 via CD3 intracellular signaling [20]. CD28 binds to B7-1 and B7-2 (CD80 and CD86) on the APC and it constitutes in naive T cells the main co-stimulatory signal. Inhibitory signals and co-stimulatory molecules such as OX40, GITR [expressed on T cells, regulatory T cells (Tregs) an DC], ICOS (expressed on activated T cells: Th1, Th2 and follicular helper T cells) are involved in regulating immune responses on the APC and T cells regulate the costimulatory process [20]. Programmed cell death protein 1 (PD-1), Cytotoxic T-lymphocyte antigen 4 (CTLA-4), T-cell immunoglobulin and mucin-domain containing-3 (TIM-3) and lymphocyte activation gene-3 (LAG-3) are types of co-inhibitory, also known as molecules that elicit an immune response. In malignant clones, there could be an exhaustion phenotype characterized by the chronic recognition of an antigen leading to feedback inhibition of effector T cell function [19]. LAG-3 is expressed in exhausted T cells and is an inhibitory receptor that constitutes a novel immunotherapeutic target with more than 20 clinical trials evaluating its possible efficacy in different tumor types [26]. A fixed-dose combination of anti-PD-1 and anti-LAG-3 has demonstrated efficacy in the treatment of unresectable or metastatic melanoma [27].

    Systemic therapy focused on immune system response in order to address and control cancer are widely known as immunotherapy and ICIs constitute one of the most relevant therapeutic approaches. The most studied ICIs are antagonistic antibodies that inhibit CTLA-4, LAG3, PD-1 and PD-L1. PD-1 and CTLA-4 suppress immune responses in different ways and the mechanisms of inhibitions in T cell responses are different: (1) PD-1 is upregulated following antigen presentation and PD-1 is a marker for T-cell activation in physiological immune responses which are different from tumor-associated T-cells that express, in later stages, PD-1 and other immune checkpoint molecules. (2) CTLA 4 is expressed early in the activation of T cells and its ligands are usually APCs in secondary lymphoid organs. Therefore, PD-1 and CTLA-4 have different roles repressing progressive immune responses [28]. Table 1 summarizes the ICIs approved by the European Medicines Agency (EMA) for different tumors and indications [2, 2988].

    List of EMA-approved immune checkpoint inhibitors with their approved indication

    ICIsAuthorApproved indicationSpecial considerationPredictive biomarker
    Anti-PD-1NivolumabLarkin et al. [29]Advanced melanomaN/AN/A
    Weber et al. [30]Resected melanoma with metastatic lymph nodes or resected metastasesAdjuvant settingN/A
    Forde et al. [31]Resectable NSCLC with high recurrence riskNeoadjuvant setting in combination with platinum-based chemotherapyPD-L1 ≥ 1%
    Borghaei et al. [32]Locally advanced or metastatic NSCLCSubsequent therapy after chemotherapyN/A
    Motzer et al. [33]Advanced renal cell carcinomaSubsequent treatmentN/A
    Choueiri et al. [34]Advanced renal cell carcinomaFirst line therapy in combination with cabozantinibN/A
    Ferris et al. [35]Recurrent or metastatic squamous head and neck cancerSubsequent therapy after platinum-based chemotherapyN/A
    Bajorin et al. [36]Resected urothelial carcinoma with high recurrence riskAdjuvant settingPD-L1 ≥ 1%
    van der Heijden [37]Locally advanced or metastatic urothelial carcinomaFirst line therapy combined with cisplatin and gemcitabineN/A
    Sharma et al. [38]Locally advanced or metastatic urothelial carcinomaSubsequent therapy after platinum-based chemotherapyN/A
    Kelly et al. [39]Esophageal or gastroesophageal junction carcinoma with residual pathologic disease after neoadjuvant chemoradiationAdjuvant settingN/A
    Doki et al. [40]Advanced unresectable, recurrent or metastatic squamous cell esophageal carcinomaFirst line therapy combined with fluoropyrimidine and platinum-based chemotherapyPD-L1 ≥ 1%
    Kato et al. [41]Advanced unresectable, recurrent or metastatic squamous cell esophageal carcinomaSubsequent therapy after platinum and fluoropyrimidine-based chemotherapyN/A
    Janjigian et al. [42]Advanced or metastatic gastric, gastroesophageal junction or esophageal adenocarcinomaFirst line therapy combined with fluoropyrimidine and platinum-based chemotherapy- HER2 negative
    - PD-L1 + with CPS ≥ 5
    PembrolizumabLuke et al. [43]Completely resected melanoma stage IIB, IIC or IIIAdjuvant settingN/A
    Schachter et al. [44]Irresectable or metastatic melanomaN/AN/A
    Reck et al. [45]Metastatic NSCLCFirst line therapy- PD-L1 + CPS ≥ 50
    - EGFR and ALK negative
    Gandhi et al. [46]Metastatic NSCLC and non-squamous cell carcinomaFirst line therapy in combination with pemetrexed and platinum-based chemotherapyEGFR and ALK negative
    Paz-Ares et al. [47]Squamous NSCLCFirst line therapy in combination with paclitaxel and platinum-based chemotherapyN/A
    Herbst et al. [48]Locally advanced or metastatic NSCLCIf EGFR or ALK positive, targeted therapy should be received prior to pembrolizumabPD-L1 + CPS ≥ 1
    Bellmunt et al. [49]Locally advanced or metastatic urothelial carcinomaSubsequent therapy after prior platinum-based chemotherapyN/A
    Balar et al. [50]Locally advanced or metastatic urothelial carcinomaUnfit for platinumPD-L1 + CPS ≥ 10
    Burtness et al. [51]Recurrent or metastatic squamous head and neck cancerFirst line therapy in combination with 5-fluorouracil + platinumPD-L1 + CPS ≥ 1
    First line therapy, monotherapyPD-L1 + CPS ≥ 50
    Choueiri et al. [52]Resected renal cell carcinoma (including resected metastases)Adjuvant settingN/A
    Rini et al. [53]Advanced renal cell carcinomaFirst line therapy in combination with axitinibN/A
    Motzer et al. [54]Advanced renal cell carcinomaFirst line therapy in combination with lenvatinibN/A
    André et al. [55]Metastatic colorectal cancerFirst line therapy or after previous chemotherapy based on fluoropyrimidinesDeficient in mismatch repair (dMMR)
    Marabelle et al. [56]Recurrent or advanced endometrial carcinomaSubsequent therapy in progression to platinum-based chemotherapydMMR
    Metastatic gastric cancerSubsequent therapy
    Metastatic small intestine cancer
    Metastatic biliary tract cancer
    Sun et al. [57]Metastatic esophageal cancer or adenocarcinoma of gastroesophageal junctionFirst line therapy in combination with platinum and fluoropyrimidine-based chemotherapyPD-L1 + CPS ≥ 10
    Schmid et al. [58]Localized triple negative breast cancerPerioperative settingN/A
    Cortes et al. [59]Metastatic triple negative breast cancerFirst line therapy in combination with chemotherapyPD-L1 + CPS ≥ 10
    Makker et al. [60]Advanced or recurrent endometrial carcinomaSubsequent therapy after platinum-based chemotherapyN/A
    Monk et al. [61]Recurrent or metastatic cervical cancerIn combination with or without bevacizumabPD-L1 + CPS ≥ 1
    Janjigian et al. [62]Unresectable or metastatic gastric or gastroesophageal junction adenocarcinoma HER2 positiveFirst line therapy in combination with trastuzumab + platinum-based chemotherapy and fluoropyrimidines- HER2 positive
    - PD-L1 + CPS ≥ 1
    Rha et al. [63]Unresectable or metastatic gastric or gastroesophageal junction adenocarcinomaFirst line therapy in combination with platinum-based chemotherapy and fluoropyrimidines- HER2 negative
    - PD-L1 + CPS ≥ 1
    Kelley et al. [64]Unresectable or metastatic biliary tract cancerFirst line therapy in combination with cisplatin and gemcitabineN/A
    DostarlimabMirza et al. [65]Advanced endometrial cancer (new diagnosed or relapse)In combination with carboplatin and paclitaxeldMMR
    Oaknin et al. [66]Primary advanced or recurrent endometrial cancer (relapse or progression after first line therapy)MonotherapydMMR
    CemiplimabMigden et al. [67]Metastatic or locally advanced squamous cell skin cancerN/AN/A
    Stratigos et al. [68]Locally advanced or metastastic basal cell carcinomaSubsequent therapy, in progression or those who not tolerate a Hedgehog pathway inhibitorN/A
    Sezer et al. [69]Locally advanced NSCLC not candidates for chemoradiotherapy or metastatic NSCLCFirst line therapy, monotherapy- PD-L1 ≥ 50%
    - No EGFR, ALK or ROS1 mutations
    Gogishvili et al. [70]Locally advanced NSCLC not candidates for chemoradiotherapy or metastatic NSCLCFirst line therapy, in combination with platinum-based chemotherapy- PD-L1 ≥ 1%
    - No EGFR, ALK or ROS1 mutations
    Tewari et al. [71]Metastatic or recurrent cervical cancerSubsequent therapy in progression to a platinum-based chemotherapyN/A
    RetifanlimabLakhani et al. [72]Locally advanced or metastatic Merkel cell carcinomaFirst line therapyN/A
    TislelizumabShen et al. [73]Unresectable, locally advanced or metastatic esophageal squamous cell carcinomaSubsequent therapy, after progression to platinum-based chemotherapyN/A
    Anti-PD-L1AtezolizumabPowles et al. [74]Locally advanced or metastatic urothelial carcinomaSubsequent therapy after platinum-based chemotherapy or first line for unfit for platinumN/A
    Rittmeyer et al. [75]Locally advanced or metastatic NSCLCAfter previous chemotherapy. If EGFR or ALK mutation, targeted therapy must have been received prior to atezolizumabN/A
    AvelumabD’Angelo [76]Metastatic Merkel cell carcinomaN/AN/A
    Powles et al. [77]Locally advanced or metastatic urothelial carcinoma with no progression after platinum-based chemotherapyFirst line therapy-maintenance therapyN/A
    Motzer et al. [78]Advanced renal cell carcinomaFirst line therapy combined with axitinibN/A
    DurvalumabAntonia et al. [79]Unresectable and locally advanced NSCLC with no progression after chemoradiotherapyMaintenance therapy after chemoradiotherapyPD-L1 ≥ 1%
    Paz-Ares et al. [80]Metastatic SCLCFirst line therapy in combination with carboplatin/cisplatin and etoposideN/A
    Burris et al. [81]Irresectable or metastatic biliary tract carcinomaFirst line therapy in combination with gemcitabine and cisplatinN/A
    Abou-Alfa et al. [82]Advanced or unresectable hepatocellular carcinomaFirst line therapy, in monotherapyN/A
    Anti-CTLA-4IpilimumabHodi et al. [83]Metastatic melanomaN/AN/A
    Anti-CTLA-4 combined with another ICINivolumab + ipilimumabLarkin et al. [2]Metastatic melanomaN/AN/A
    Motzer et al. [84]Metastatic renal carcinoma with intermediate-high riskFirst line therapyN/A
    Baas et al. [85]Unresectable malignant pleural mesotheliomaFirst line therapyN/A
    Kato et al. [86]Advanced unresectable, recurrent or metastatic squamous cell esophageal carcinomaFirst line therapyPD-L1 ≥ 1%
    Lenz et al. [87]Metastatic colorectal carcinomaSubsequent therapy after fluoropyrimidine-based chemotherapydMMR
    Tremelimumab + durvalumabAbou-Alfa et al. [82]Advanced or unresectable hepatocellular carcinomaFirst line therapyN/A
    Tremelimumab + durvalumab + platinum-based chemotherapyJohnson et al. [88]Metastatic NSCLCFirst line therapyNo EGFR or ALK mutations
    Display full size

    ICIs: immune checkpoint inhibitors; PD-1: programmed cell death protein 1; PD-L1: programmed death ligand 1; NSCLC: non-small cell lung cancer; HER2: human epidermal growth factor receptor 2; CPS: combined positive score; EGFR: epidermal growth factor receptor; ALK: anaplastic lymphoma kinase; ROS1: ROS proto-oncogene 1 receptor tyrosine kinase. N/A: not applicable

    CTLA-4

    CTLA-4 functions by being expressed on the surface of CD4+ and CD8+ T cells, where it has a stronger binding affinity for the costimulatory molecules CD80 and CD86 (B7-1 and B7-2) on APCs compared to the T cell costimulatory receptor CD28 [89].

    Naive T cells require a stimulation signal through the TCR, derived from an antigen, and a subsequent costimulation signal which passes when the CD28 on T cells engage with B7.1/CD80 or B7.2/CD86 on APCs [9092]. CTLA-4 is an inhibitory receptor present in T cells that decreases T cell activity and it can be increased when a T cell is activated [9395]. CTLA-4 can decrease the costimulation process by binding and competing with a higher affinity for the same ligands as CD28, therefore, inhibiting T cell activity [96, 97]. CTLA-4 can suppress T cell activity via the extrinsic and intrinsic pathways. In the extrinsic inhibition, CTLA-4 depresses the optimal level of costimulatory signals required by the T cell by competing with CD28 for ligands [98]. In the intrinsic inhibition, CTLA-4 recruits phosphatases and transcription factors (NFAT, NF-κB and AP-1) are also inhibited, which are associated with the activation of T cells [99105]. On the other hand, Tregs express CTLA-4 on the surface which control the generation and function of Tregs [106110], thus, suppressing immune responses.

    One of the main ways anti-CTLA-4 functions is by enabling macrophages to eliminate Treg cells from the tumor microenvironment (TME) [89]. The effectiveness of anti-CTLA-4 antibodies relies, at least partially, on reducing tumor-infiltrating Tregs through interactions with human Fcγ receptors (FcγRs) and human immunoglobulin Gs (IgGs). Enhancing antibody-dependent cell-mediated cytotoxicity—either by optimizing the fragment crystallizable (Fc) region or by the presence of high-affinity FcγR variants—boosts therapeutic efficacy, but primarily in highly immunogenic tumors [89]. Anti-CTLA-4 immunotherapy does not eliminate FOXP3+ cells in human tumors, indicating that its effectiveness could be improved by altering the Fc regions of monoclonal antibodies to enhance Fc-driven depletion of Tregs within the tumor [111]. Depleting both intratumoral and nodal Tregs independently of CTLA-4 led to even stronger antitumor effects, indicating that targeting nodal Tregs could be a valuable approach to boosting the effectiveness of current immunotherapies [112].

    Anti-CTLA-4 antibodies’ development made the ICIs development possible. The first evidence of the antitumor effects of anti-CTLA-4 activity was in 1996 when mice with colon carcinoma cells treated with anti-CTLA-4 antibodies obtained an objective tumor response [113], and also a protective immunity for the long term was observed. In 2010, the survival benefit of anti-CTLA-4 antibody ipilimumab was demonstrated in a phase III clinical trial over a melanoma-specific peptide vaccine in patients with melanoma.

    PD-1

    PD-1 is an immunoreceptor that has two tyrosine residues in the cytoplasmic regions and belongs to the immunoglobulin superfamily [114]. It is a transmembrane protein that regulates programmed T cell death [115] expressed on macrophages, Langerhans cells, dendritic cells (DCs), B cells and T cells, but its expression is highly increased on exhausted T cells [116, 117]. Hematopoietic and tumor cells can express PD-L1 on the surface, whereas PD-L2 is more often present in hematopoietic cells [118, 119]. Monocytes, DCs and NK cells can express PD-1 and/or PD-L1. PD-1 has an inhibitory activity that engages to PD-L1 (B7-H1) and PD-L2 (B7-H2) which downregulates T cell immune responses [118, 119]. Thus, the engagement of PD-1 and PD-L1/2 lead to phosphorylation of its cytoplasmic immunoreceptors and inhibits tumor cell apoptosis, contributes to peripheral T effector cell exhaustion and the transformation of T effector cells to Treg cells [7, 120]. The specific phosphorylation of the immunoreceptor tyrosine-based switch motif (ITSM) recruits the Src homology region 2 domain-containing phosphatase-2 (SHP-2) and represses the activity of some intracellular molecules that propagate signals following the TCR, therefore, mediates the PD-1 inhibitory functions [28, 114]. The dephosphorylation events dull the inflammatory reaction of the affected cells: (1) in T cells, PD-1 signals dephosphorylate ZAP70 and CD3ζ, which transduce signals downstream of TCR engagement [114]; (2) in B cells the recruitment of SHP-2 dephosphorylates effector molecules such as Syk and PI3K [121]. The Ras/MEK/ERK pathway, that controls cell proliferation, survival and growth, can be inhibited via SHP-2 which may contribute to a restrained proliferation after activation [122]. The second key pathway of PD-1 intracellular signaling in T-cells involves enhancing the expression of E3 ubiquitin ligases at the transcriptional level [123]. It has been shown that PD-1 works together with LAG-3 to suppress T-cell activity via the E3 ubiquitin ligase pathway, which plays a crucial role in resistance to PD-1/PD-L1 blockade monotherapies [124]. The function of the PD-1/PD-L1/PD-L2 axis is summarized in Figure 1.

    The function of the PD-1/PD-L1/PD-L2 axis. This illustration highlights the PD-1/PD-L1/PD-L2 axis as a critical immune checkpoint in oncology. PD-1, expressed on T-cells, binds to PD-L1 (often overexpressed on tumor cells) or PD-L2 (on tumor cells or antigen-presenting cells), triggering SHP-2 to inhibit TCR and CD28 signaling, thus suppressing T-cell activation and promoting immune evasion. This is a key mechanism of resistance to immunotherapy, as tumors can upregulate PD-L1 in response to IFN-γ from the microenvironment. PD-1: programmed cell death protein 1; SHP-2: Src homology region 2 domain-containing phosphatase-2; TCR: T cell receptor; PD-L1: programmed death ligand 1; IFN-γ: interferon-gamma

    Targeting PD-1/PD-L1 has been evaluated in order to boost antitumor activity [125130]. In mice, the interplay between PD-L1 present in tumor cells and PD-1 present on cytotoxic T cells resulted in an accelerated tumor growth, thus, PD-L1 antibody therapy obtained a contributed to tumor progression [125]. Furthermore, PD-L1 and PD-L2 expression has also been associated to poor disease prognosis in various cancer types [131135]. Numerous studies have demonstrated the efficacy of anti-PD-1 and anti-PD-L1 antibodies as a systemic therapy for cancer. Blocking PD-L1 inhibits the PD-1/PD-L1 axis as well as the PD-L1/CD80 cis interplay on DCs which ends up liberating more CD80 molecules that are able to increase T cell priming [136]. Ongoing clinical trials are currently investigating the use of monoclonal antibodies targeting other immune checkpoints, such as TIM-3, LAG3, and T-cell Immunoreceptor with Ig and ITIM domains (TIGIT), in combination with PD-1 inhibitors. For instance, studies on TIM-3 have shown that blocking both PD-1 and TIM-3 together significantly boosts the immune activity of effector T cells in animal models of diseases like acute myeloid leukemia and lung cancer, achieving greater effectiveness than PD-1 inhibitors alone [137140]. Atezolizumab, avelumab and durvalumab are the three main anti-PD-L1 antibodies that have been approved for various cancer types and, although all of these antibodies stop the interaction of PD-L1 with PD-1 and CD80, there are some differences in their mechanisms of action that should be noted. The Fc region has a modification in durvalumab and atezolizumab that prevents exhaustion of T cells that express PD-L1 by the removal of antibody-dependent cellular cytotoxicity. Whereas, avelumab contains the native Fc region that induces antibody-dependent cellular cytotoxicity through the engagement of FCγ receptors on NK cells [28].

    PD-L1

    PD-L1 is part of the B7 family of molecules that regulate immune responses, acting as either co-stimulatory or co-inhibitory signals. It is expressed by various cell types, including tumor cells [141]. PD-L1 is a membrane-bound protein composed of an immunoglobulin-like extracellular region, a transmembrane segment, and a short intracellular domain and can also bind CD80 as PD-1 [142]. PD-L1 plays a crucial role in preserving peripheral tolerance and supporting DCs in presenting antigens to T cells. In cancer, its expression in tumors is strongly associated with advanced disease and poor outcomes, often signaling resistance to conventional therapies such as chemotherapy and radiotherapy [141, 143]. One of the ways PD-L1 exerts its protective effects is by inhibiting IFN-gamma signaling within cancer cells through its intracellular domains [143]. This also enhances cancer cell resistance mechanisms by disrupting the JAK/signal transducer and activator of transcription (STAT) signaling pathways through mutations [144].

    The signaling pathway that drives the adaptive expression of PD-L1 and PD-L2 in response to IFNs is crucial for advancing PD-1 blockade therapies in cancer treatment. When T cells recognize tumor antigens, they release IFNs, which stimulate cancer cells or other cells in the TME to express PD-L1. This suppresses the antitumor immune response in a process called adaptive immune resistance. This mechanism specifically limits T-cell recognition of cancer cells while preserving immune responses to other antigens, preventing widespread immune suppression [145, 146].

    PD-L2

    PD-L2 (also known as B7-DC or CD273) is a type I transmembrane protein belonging to the B7 family. Its extracellular region consists of an immunoglobulin (Ig)-like V-type domain and an Ig-like C2-type domain [147]. PD-L2 is found not only in tumor cells but also in immune cells, and its elevated expression has been shown to play a key role in cancer development and immune evasion. PD-L1 and PD-L2 are ligands of the PD-1 immune checkpoint, which suppresses immune responses. Abnormal PD-L2 expression plays a major role in cancer development and progression by helping tumor cells evade detection and destruction by the immune system [147]. Their expression can be triggered in tumors by IFN exposure, allowing cancer cells to evade immune attack. This mechanism plays a crucial role in immunotherapies that target PD-1 inhibition. When PD-1 binds to PD-L2, it significantly suppresses TCR-driven proliferation and cytokine release in CD4+ T cells. At low antigen levels, PD-L2-PD-1 interactions weaken the strong signaling from B7-CD28. However, when antigen levels are high, these interactions limit cytokine production but do not prevent T cell proliferation [119].

    LAG-3

    LAG-3 is a protein found on B cells, certain T cells, NK cells, and tumor-infiltrating lymphocytes (TILs), playing a role in regulating immune checkpoint pathways [148]. LAG-3 is a membrane protein that interacts with MHC class II, promoting Treg function while suppressing T cell proliferation, activation, and balance. Similarly, Programmed Death 1 (PD-1) inhibits T cell activity. Both LAG-3 and PD-1 are temporarily expressed on CD8 T cells following acute stimulation [149]. It enhances the activity of Tregs by interacting with MHC class II molecules, which suppresses T cell differentiation and proliferation [150].

    LAG-3 expression on T cells helps to suppress activation and maintain immune balance [151]. Earlier research showed that transgenic CD4 T cells exposed to their specific self-antigen in vivo exhibited increased LAG-3 expression and became anergic. In this model, these tolerized CD4 T cells demonstrated regulatory functions both in vitro and in vivo, and their in vitro suppressive activity was blocked using a LAG-3-specific monoclonal antibody [152]. Naive CD8 T cells initially express low levels of LAG-3, but its expression sharply rises in response to antigen stimulation [153]. Blocking LAG-3 with specific antibodies can restore the function of exhausted T cells, improving their ability to target and destroy tumor cells.

    The simultaneous increase in PD-1 and LAG-3 expression on CD4 and CD8 T cells is a key factor contributing to resistance against treatments that target PD-1 or PD-L1 alone [154, 155]. The presence of both PD-1 and LAG-3 on TILs has been proposed as a marker of impaired immune function in non-small cell lung cancer (NSCLC) [156]. An increasing amount of research suggests that the co-expression of PD-1 and LAG-3 in T cells contributes to resistance against anti-PD-1 and anti-PD-L1 treatments [157160].

    Relatlimab (a human IgG4 LAG-3-blocking antibody) combined with nivolumab (anti-PD-1) has been proven to be a safe and effective treatment for advanced melanoma [27]. Analysis of biospecimens from an ongoing study showed that relatlimab plus nivolumab treatment enhanced CD8+ TCR signaling and modified CD8+ T cell differentiation, boosting cytotoxic activity while maintaining characteristics of exhaustion [155]. The co-expression of cytotoxic and exhaustion markers was regulated by PRDM1, BATF, ETV7, and TOX. Furthermore, effector function increased in clonally expanded CD8+ T cells that appeared after treatment with the combination [155].

    Mechanism of resistance to ICI

    Tumor intrinsic mechanism

    Neoantigen exhaustion

    One of the main contributors to primary resistance to ICIs associated with tumor intrinsic mechanisms is the low availability of neoantigens and low mutational load [161]. Neoantigens are tumor-specific antigens that can derive from tumor genome instability that has previously led to somatic mutations in different cancer types [161]. Tumors can acquire a high neoantigen load through mismatch repair or genomic instability or, as observed in NSCLC and melanoma, the mutation acquisition could be through carcinogen-induced DNA damage or UV-damage, respectively [11, 162, 163]. Neoantigens are able to confer high immunogenicity with more affinity to MHC-II which then derives to a more intense immunological response with an increased antitumor T cell response with ICIs [164, 165]. ICIs can activate tumor infiltrating T cells which are more abundant in patients with mutations in DNA repair as MSH2, MLH1 and ATM [166, 167]. Neoantigen depletion can be associated with primary resistance to ICIs, and in the presence of tumor immune elimination, tumor cells can evade the antitumoral response by the HLA loss of heterozygosity or enhance the loss of tumor-specific neoantigens [161]. Therefore, tumor response to ICIs could fail if there is tumoral neoantigen depletion [168].

    Disruption of critical signaling pathways

    IFN-γ is a cytokine that deploys proapoptotic and antriproliferative effects, induces the boost of the MHC I in tumor cells and, furthermore, promotes CD8+ cytotoxic T cell activity uplifting PD-L1 levels, which all together accounts for the interplay of IFN-γ in the initiation and maintenance of antitumor response [169, 170]. Effector T cells release IFN-γ which promotes JAK-STAT signaling cascades, IFN-γ binds to the IFN-γ receptor it activates the receptor-associated kinases JAK1/2 and STAT1/2, that promote tumor cell death by upregulation MHC class I expression [161, 171173]. PD-L1 functions as an anti-apoptotic signaling molecule in cancer cells by interfering with type I and II IFN signaling pathways, helping tumor cells resist immune-mediated cell death [174, 175]. Tumors who harbor mutations in IFNGR1/2 and JAK1/2 have been identified in non-responders to anti-PD-1 and anti-CTLA-4 ICIs [176, 177]. Therefore, the dysregulation of the IFN-γ pathway in tumor cells are associated with a subdued response to ICIs and the loss of function alterations in the JAK/STAT pathway result in the loss of MHC class I by inducing cell resistance to IFN [69]. In this context, an IFN-γ signature has demonstrated a predictive value in cancer patients treated with anti-PD-1 [178180]. Inflammatory cytokines and tumor-promoting inflammation, such as IL-6, IL-8 and C-reactive protein, have been linked to ICI resistance and can play a role as potential biomarkers [181]. Ceramide metabolism alterations have also been associated with TNF-induced melanoma cell dedifferentiation and certain ceramide metabolites could be predictive biomarkers of immunotherapy resistance [182].

    Defects in antigen presentation

    The recognition of antigens on MHC by APC is one of the mainstays of T cell activation in immunological responses [161]. β2M is a gene involved in the modulation of antigen presentation to cytotoxic CD8+ T cells by MHC class I complex [183185] and it restores MHC-I/peptide complex formation [186]. β2M loss of function has been associated with MHC class I deficiency in cancer cells which can confer resistance from immune surveillance by reducing T cell recognition [186]. Cancer patients with a β2M loss show dysregulated immune responses because of the impairment of surface MHC class I via the degradation of heavy chain of MHC class I in cytosol. Another gene involved in antigen presentation is MEX3B, which downregulates MHC I expression in tumors, has been observed at higher levels in non-responders to anti-PD-1 cancer patients compared to responders [187].

    The expression of PD-L1

    PD-L1 expression on tumor cells is a well-known biomarker that has been associated with response to ICIs [185187]. Previously, we have noted the importance of antigen presentation and activated CD8+ T cell responses in the response of ICIs, thus, PD-L1 expression associates with tumoral immune activation at this level [186]. Therefore, PD-L1 low-expression in patients with NSCLC and melanoma has shown up to 10% of response rates compared to 40–50% response rates in patients with increased PD-L1 expression [185, 186]. Nonetheless, anti-PD-1 treatments have shown to be effective in PD-L1 negative tumors, and on the other hand, patients with an increased PD-L1 expression don’t always respond to ICIs [6, 11].

    Tumor microenvironment

    TME compromises non-immune stromal cells (fibroblasts and endothelial cells) and immune cells which includes B cells, T cells, NK cells, MDSCs and DCs [188]. ICIs response may be altered depending on the TME [161].

    Anti-tumor response of CD8+ T cells

    As previously mentioned, ICIs seek to promote anti-tumor immune responses by T cells [189, 190] and the amount of CD8+ T cells in the TME have been related with an overall increased response to PD-1/PD-L1 blockade [6, 191]. In this setting, the amount of T cells in the TME is crucial for the ICI response [161], but the mere presence is not enough to predict response in patients treated with ICIs [188]. There is an existing high variability of tumor-infiltrating T cells across tumor types [192200] and by single-cell RNA sequencing differences between lymphocytes have been observed: (1) cytotoxic genes as GZMA has been related with PD-1 blockade response [201]; (2) overall, gene signatures related to T cell cytotoxic activity have been associated with anti-CTLA-4 ipilimumab response [202]. Tissue-resident T cells and new infiltrating T cells can be found in the TME and response to ICIs vary depending on their cytotoxic effect [188, 203]. Evidence suggests that ICIs promote that less-differentiated memory-like CD8+ T cells convert into effector CD8+ T cells, enhancing anti-tumor response [188].

    Furthermore, CD8+ T cells play a role in the genesis of immunotherapy adverse events, for example, gastrointestinal adverse events. Future therapeutic strategies based on blocking the JAK pathway could be beneficial in this context and modulate immunotherapy adverse events [204]. Tofacitinib is a pan-Janus kinase inhibitor that has shown to control immunorelated colitis in case reports and small case series, warranting prospective validation [205].

    The activation of the WNT/β-catenin pathway downregulates T cell activation and has been associated with a paucity of T cell infiltration in metastatic melanoma [161, 206208]. The mechanism through which the activation of β-catenin suppresses ICIs can be related to the inhibition of CCL4 transcription, described in mouse models, which results in impaired CD8+ T cell infiltration [19]. On the contrary, CCL4 levels can recover when there is no activation of β-catenin, which facilitates CD8+ T cells response to ICIs [161]. Moreover, PTEN deficient cells are uncapable of IFN signaling pathway activation and that has also been associated with a decrease in the immune response of the TME [209, 210]. PTEN deficiency can also increase T cell suppression in the TME by the increase of downregulating factors and decreasing the destructive functions of cytotoxic T lymphocytes (CTLs) [209, 210].

    Anti-tumor response of CD4+ T cells

    CD4 T cells play a role in cancer immunity by providing essential helper functions. Additionally, studies have shown that they can also exhibit direct cytotoxic activity in certain cancer patients [211]. Cancer immunotherapy research has largely centered on CD8+ T cells within the TME [116]. However, studies suggest that effective antitumor immunity requires tumor cells to present MHC class II-binding neoantigens, which are recognized by CD4+ T cells [212]. CD4+ T cells likely play a crucial role in sustaining the cancer immunity cycle by ensuring a continuous supply of CTLs to the TME [213].

    CD4+ T cells develop into distinct functional subtypes [214]: Th1 cells, which enhance CD8+ T cell responses to eliminate intracellular pathogens, and Th17 cells, which recruit neutrophils to combat fungi and extracellular bacteria. In cancer immunity, Th1 cells are considered key players by producing IFNγ and boosting CD8+ T cell activity. Meanwhile, Th17 cells, known for their stem cell-like characteristics, are thought to support long-lasting antitumor immunity [215].

    Zuazo et al. [154]. showed that baseline systemic CD4 immunity serves as a key factor in determining clinical response. All patients who showed objective treatment responses had functional systemic CD4 T cells, which could be identified prior to therapy by their high proportion of memory CD4 T cells. Inomata et al. [216]. indicated that the proportion of peripheral CD45RA CD4+ T cells was linked to progression-free survival after beginning ICI therapy, independent of multiple clinical variables. Also, neoantigen-specific CD4 T cells are fundamental [217].

    Other lymphoid structures and B cells

    PD-1 expression can also be found in regulatory B cells [218] or CTLA [219] and ICIs could have a direct repercussion on B cells. B cell activation and tertiary lymphoid structures have been correlated with tumor response to ICIs in several types of tumors [161, 188]. In melanoma, single-cell studies have observed that a higher number of B cells in the TME were related to an increased tumor response to PD-1 blockade [192] and there is a higher number of memory-like B cells and plasmablast-like cells in responders [220]. High levels of B-cell-secreted antibodies were found in melanoma anti-CTLA-4 or anti-PD-1 responders [221]. Germain et al. [222] observed that patients with lung cancer that had an infiltration of B-cell in tertiary lymphoid structures which served as a defensive immunological marker.

    Macrophages, dendritic cells and NK cells

    These other immune cells have an impact on tumor progression and can also be present in the TME [188]. Macrophages show two main states of polarization: (1) M1 macrophages which are activated and can promote CD8+ T cells by antigen presentation and the release of cytokines; (2) activated M2 macrophages secrete TGF-β and show pro-angiogenic and immunosuppressive properties [223]. Furthermore, activated M2 macrophages have been related with hyperprogression in NSCLC patients treated with anti-PD-L1 blockade and, the increase in M2 phenotype could be a consequence of the union of ICIs to macrophage Fc receptors [224]. The antitumor M1 phenotype, CD68+ CD16+ activated M1 macrophages, have been observed in melanoma patients who respond to CTLA-4 blockade therapy in contrast to non-responders [225]. The protumor M2 phenotype can express PD-1 more often and it has been observed that PD-(L)1-blockade could return the M2-polarized function into an antitumor M1 phenotype capable of malignant cell phagocytosis [188, 226]. Nevertheless, the M1/M2 macrophages dichotomy could not reflect other macrophage phenotypes that can be present at the TME [227]. Tumor-associated macrophages (TAMs) can promote tumor cell angiogenesis, proliferation and lymphangiogenesis, invasion and metastasis and can have an impact on anti-tumor therapy response [228, 229]. TAMs upregulate the CCL22 levels that promote Treg production in order to indirectly suppress immune function [230]. TAM subsets and subpopulations, TREM2+, SPP1+, MARCO+, FOLR2+, SIGLEC1+, APOC1+, C1QC+, among others, have been described in cancer and have been associated with poor prognosis [231]. More specifically, MARCO+ macrophages are a subset of TAMs that are immunosuppressive and the inhibition of MARCO was associated with pro-inflammatory phenotype and anti-tumoral coverage of T cells and NK cells in experimental models [232235].

    For the immune response led by CD8+ T cells to occur, DCs, among other antigen-presenting cells, present antigens and they can be divided in classical DCs (cDCs) and type I IFN-producing plasmacytoid DCs (pDCs). Moreover, cDCs are classified into cDC1, that promotes exogenous antigen presentation to CD8+ T cells, and cDC2 which induce Th-2 or Th-17 responses and favor antigen presentation to CD4+ T cells [236, 237]. cDC1 produces IL-12 which has been related with effective anti-PD-1 blockade in mouse models [77]. DCs liberate IL-12 in the presence of IFNγ secreted by T cells during anti-PD-1 blockade therapy, therefore, promoting T-cell immune responses [169].

    NK cells are innate lymphocytes that execute cytotoxic functions or secrete cytokines when there is a lack of expression of class I MHC [238, 239]. The presence of PD-1+ NK cells with an activated phenotype have been described in the TME and the PD-L1 binding has been associated with an inadequate function of the NK cells [239, 240]. NK cells exhaustion in mouse models have been observed to diminish the PD-1 and PD-L1 effects on inhibition [241].

    Extracellular matrix and TME metabolism

    Enzymes in the extracellular matrix (ECM), such as matrix metalloproteinases and a distintegrin and metalloproteinsases, can modulate the cell-cell and cell-ECM interplay [161]. Furthermore, the ECM accounts for growth factors and cytokines, in this scenario, a rigid ECM promotes hypoxia and angiogenesis with a depletion of anti-tumor responses [242244].

    Metabolism in the TME is regulated by immune and tumor cells that secrete cytokines, such as ILs, IFN and tumor growth factors, which can alter response to ICIs [161, 245247]. Glucose is a key factor for the activation of cytotoxic CD8+ T cells and cancer cells compete for glucose consumption in a TME characterized by low-glucose levels and hypoxia [161]. Furthermore, glutamine depletion promoted by cancer cells can decrease T cell activity and suppress immune response [161]. In the tumor cell surface, CD73 and CD38 promote adenosine production which suppresses CD8+ T cell function and generates an acquired resistance to anti-PD-1 blockade [248250]. Also, indoleamine 2,3 dioxygenase (IDO) can degrade tryptophan into kynurenine that can ultimately activate aryl hydrocarbon receptor that favors an immunosuppressive response by favoring T-cell differentiation to Tregs [161, 251, 252].

    Host

    The primary host-related factors influencing the effectiveness of ICI treatments are diet and the gut microbiome.

    Diet

    Nutrient consumption has the potential to directly or indirectly influence both the immune system and cancer progression. The majority of existing data are derived from human studies rather than experimental animal models [253]. While potential mechanisms have been identified that may impact immune function and, in turn, cancer growth and response to ICI, there is limited understanding of how these mechanisms affect and modify therapeutic outcomes.

    However, there is some evidence regarding the effect of the ketogenic diet on anticancer efficacy. It has been shown to amplify the effects of anti-CTLA-4 therapy by downregulating PD-L1 expression, while also increasing levels of IFN and genes involved in antigen presentation [254, 255]. Additionally, obesity and dietary restrictions have been reported to affect the responsiveness to immunotherapy, though findings in this area remain inconsistent [256]. Moreover, research has explored the immunomodulatory role of vitamin D, which impacts the adaptive immune response by activating Th1/Th17 cells, thus influencing ICI therapy [257]. Various studies have examined how different dietary patterns affect immune responses to ICIs. While some dietary approaches have been found to enhance immune activity, the exact mechanisms are not well understood, and detailed documentation on dietary influences is often lacking [161].

    Another series of nutrients have been investigated for their potential role in modulating the response to ICIs [258]. Specific dietary elements, such as vitamins, fatty acids, and other nutrients, may enhance the efficacy of anticancer immunotherapies by influencing immune responses. Caloric restriction and fasting-mimicking diets could potentially lower cancer risk and improve immunotherapy outcomes by inducing immunogenic cell death and increasing the sensitivity of tumor cells. Additionally, polyphenolic compounds such as resveratrol, apigenin, and curcumin may affect PD-L1 expression, thus modulating immune responses against cancer cells and possibly boosting the effectiveness of immune checkpoint therapies. Low-protein diets have shown varied effects on different cancers, with evidence suggesting they might significantly inhibit tumor growth by altering immune responses and amino acid metabolism.

    Furthermore, dietary habits have emerged as a major factor in modulating the gut microbiome. Research indicates that nutritional status can affect the microbiome, impacting intestinal balance and immune processes. Consumption of fruits and vegetables leads to more profound and intricate changes in the gut microbiome [259, 260].

    Future research should focus on evaluating the effects of diet, nutritional status, and gut microbiota in immunotherapy studies, clarifying the relationship between diet and circulating immune cells and/or the TME.

    Microbiota

    In recent years, extensive research has led to the identification of numerous microbial communities in the human gut and skin (estimated to be around 30 billion microorganisms per person), and changes in these microbial communities may lead to profound changes in health. Their functions include regulating the immune system [261263].

    Mouse models maintained under germ-free conditions have elucidated the pivotal influence of the gut microbiota on the ontogeny and modulation of multiple organs and physiological systems, encompassing the immune and endocrine systems, as well as the blood, liver, and lungs [264]. Within the intestinal milieu, the microbiota sustains epithelial equilibrium and facilitates the establishment of gut-associated lymphoid tissue. Furthermore, it enhances the secretion of epithelial cytokines, which subsequently orchestrate the functions of T and B lymphocytes, macrophages, and polymorphonuclear leukocytes [265, 266]. Pro-inflammatory cytokines, such as IL-1β, TNF-α, IL-2, IL-6, IL-15, IL-21, and IL-23, are capable of initiating an inflammatory cascade, whereas cytokines like IL-10 and TGF-β exert anti-inflammatory effects. The interplay between these pro-inflammatory and anti-inflammatory mediators governs whether the intestinal environment adopts an inflammatory or homeostatic profile [267]. The microbiota contributes to various immunological processes, including the maturation of DCs, T cell differentiation, and cytokine production, all of which help regulate antitumor immune responses. Moreover, the gut microbiome produces a diverse array of antimicrobial peptides (AMPs) that defend the host against various pathogens and can enhance immune responses by boosting innate immunity and reducing inflammation [161]. Certain AMPs have the ability to attract and activate immune cells and trigger inflammatory responses. This functional aspect of AMPs makes them valuable as immunomodulators, potentially explaining how the gut microbiota might improve the effectiveness of ICIs [268, 269].

    Research has shown that the gut microbiota’s composition can influence the effectiveness of ICIs. For instance, the efficacy of anti-CTLA-4 agents is impacted by specific microbiota profiles, such as those involving Bacteroides fragilis, Bacteroides thetaiotaomicron, and Burkholderiales, which can affect the TH1 immune response triggered by IL-12 [270, 271]. A study by Peng et al. [272] highlighted a connection between the gut microbiome and clinical responses to anti-PD-1/PD-L1 therapy in gastrointestinal cancers. This research found that a higher Prevotella/Bacteroides ratio (an indicator of positive regulation) is associated with better responses to anti-PD-1/PD-L1 treatments. Additionally, responders exhibited a significantly higher abundance of certain microbiota, including Prevotella, Ruminococcaceae, and Lachnospiraceae.

    Fecal microbiota transplantation (FMT), which involves transferring gut microbiota from one individual to another’s intestinal system, has emerged as a potential method for reshaping the structure and function of gut microbiomes in recipients. Extensive research has been carried out on using FMT to alter gut microbiota in various in vivo models and cancer patients, with the aim of overcoming resistance to ICIs [273275].

    Scientific research indicates that probiotics can influence both innate and adaptive immune responses. Klein and colleagues [276] found that daily probiotic supplementation notably enhanced the proportion of granulocytes and monocytes exhibiting phagocytic activity in a healthy cohort, compared to a placebo. Gill’s group [277], also confirmed these findings, noting a significant rise in serum antibody responses to both orally and systemically administered antigens in mice treated with probiotics. Lactobacillus and Bifidobacterium are among the most extensively studied probiotics in both animal models and clinical trials. Their immunomodulatory potential has been demonstrated through research on the prevention of allergic diseases [278]. Unfortunately, to date, there are no studies demonstrating their efficacy as adjuvant therapy for ICIs. Figure 2 summarizes the determinants of ICI effectiveness and strategies to enhance its antitumor activity.

    Determinants of ICI effectiveness and strategies to enhance its antitumor activity. The factors influencing ICI effectiveness can be grouped into: (a) Tumor cell-intrinsic mechanisms, including 1. loss of neoantigens, 2. deficiencies in antigen presentation, 3. disruptions in interferon signaling; (b) the TME; (c) host-related factors, such as 1. nutritional status, 2. the gut microbiome; (d) the constraints of monotherapy. To address these, strategies to enhance ICI antitumor activity include: (a) employing NK cell-based therapies and oncolytic virotherapy targeting tumor cell-intrinsic mechanisms; (b) using pharmacological inhibitors, cytokines, interleukins, and adjuvants to counteract immunosuppression within the TME; (c) implementing dietary interventions and FMT; (d) adopting combination therapy approaches. ICI: immune checkpoint inhibitors; TME: tumor microenvironment

    Discussion

    Probably, one of the main challenges for the future of oncology will be how to reverse both primary and acquired resistance to immunotherapy. One of the most significant attributes of ICIs is their ability to induce a durable response. Unfortunately, this outcome is only observed in a subset of cancer types and specific patients, owing to the issue of ICI resistance.

    Despite the rapid progress in understanding drug resistance to immunotherapy, clinical trials focused on overcoming ICI resistance still face significant challenges. With advancements in medical science, the emphasis has shifted towards ‘personalized medicine’ and ‘precision treatment’ in oncology. Biomarkers play a crucial role in the detection, management, and prognosis of tumors. Currently, PD-L1 remains the most frequently utilized biomarker to forecast immunotherapy effectiveness, though it has notable limitations. Additionally, other indicators such as tumor mutational burden, TILs, and microsatellite instability are also valuable in predicting the success of immunotherapeutic approaches.

    Consequently, there is a pressing need to further elucidate the mechanisms underlying resistance to ICIs. This review has examined potential mechanisms of resistance involving tumor cells, the TME, and host-related factors. Additionally, resistance mechanisms may occur concurrently or sequentially. Given the intricate nature of these mechanisms and the TME, it is essential to explore combined strategies to address ICI resistance. Currently, a standardized approach to this issue remains elusive. Integrating immunotherapy with other modalities such as chemotherapy, radiotherapy, and targeted therapy represents a viable strategy for managing a range of cancers.

    The following sections briefly discuss different research strategies to overcome ICI resistance (Table 2):

    Immune checkpoint inhibitor combination strategies

    CombinationEffects
    ICI + ICIPromote T cell activation and the immune response to tumor cells
    ICI + TTIncrease T-cell infiltration and IFN-gamma expression, besides its specific action on the target
    ICI + ChTReduce the number and activity of immune-suppressive cells and stimulate tumor-specific immune responses by inducing the immunogenic cell death
    ICI + RTICI enhances the immunogenic effects of RT and RT reduces the number and activity of immune-suppressive cells and remodeling of the TME by secreting inflammatory factors
    ICI + AABoost immune cell infiltration, elevate adhesion molecules, promote antigen presentation and reduce Treg immersion
    ICI + ACTIncrease IFN-gamma-expression
    ICI + nanomedicineTransform “deserted tumors” to “inflamed tumors” by boosting T-cell priming and immersion
    Display full size

    ICI: immune checkpoint inhibitor; TT: targeted therapy; IFN: interferon; ChT: Chemotherapy; RT: radiotherapy; TME: tumor microenvironment; AA: anti-angiogenesis; ACT: adoptive cellular therapy

    Combining ICIs

    The co-administration of different ICIs has shown to enhance immune responses through their complementary effects. Specifically, agents targeting CTLA-4 and PD-1/PD-L1 operate through distinct signaling pathways. Numerous clinical trials have demonstrated that dual inhibition of PD-1/PD-L1 and CTLA-4 yields superior efficacy compared to monotherapy. Ongoing clinical studies are exploring the efficacy of ICIs in combination with inhibitors targeting additional immune checkpoints, such as TIM-3, LAG-3, V-domain Ig suppressor of T-cell activation (VISTA) and TIGIT [279, 280].

    Combination of ICI with targeted therapy

    Clinical trials have demonstrated that integrating ICIs with targeted therapies can enhance treatment outcomes. For instance, the addition of atezolizumab to targeted therapy has been shown to improve PFS in melanoma patients with BRAF mutations [281]. Similarly, combining niraparib with pembrolizumab has resulted in higher response rates among individuals with metastatic BRCA-mutant triple-negative breast cancer [282]. However, the effectiveness of this combined approach is currently limited to specific tumor types.

    Combination of ICI with chemotherapy

    A multitude of studies have explored the effectiveness of combining ICIs with chemotherapy. This approach not only reduces the number of immunosuppressive cells within the TME but also increases the presentation of tumor antigens by inducing tumor cell apoptosis [283, 284]. Such treatment enhances tumor cell clearance and boosts immune system recognition through improved immune surveillance. Presently, extensive research is being conducted on the concurrent and sequential use of ICIs with chemotherapy, revealing significant efficacy improvements across a range of cancer types [285].

    Combination of ICI with radiotherapy

    Radiotherapy induces cancer cell apoptosis through DNA damage and influences immunogenicity by releasing inflammatory mediators, which can elevate neoantigen expression. It also significantly impacts the TME by secreting these inflammatory factors [286289]. Nonetheless, challenges remain in optimizing the balance between enhancing immune stimulation and reducing immunosuppressive effects, as well as in managing or preventing potential increases in toxicity, such as pneumonitis.

    Combination of ICI with anti-angiogenesis agents

    Tumor angiogenesis is partially regulated by the immune-suppressive environment within the tumor. The activation of T cells results in the production of IFN-γ, which interacts with its receptor on endothelial cells in tumors, leading to the regression of tumor-associated blood vessels [290, 291]. Evidence from murine models shows a synergistic effect between ICIs and anti-angiogenic agents [292, 293]. These agents inhibit the development of blood vessels essential for tumor growth and metastasis and help to restore the TME [292, 294]. In vivo studies have reported an increase in anti-tumor immune cell populations and a decrease in pro-tumor immune cells [295]. Therapies that combine ICIs with anti-angiogenic agents have demonstrated greater efficacy compared to monotherapy [296, 297].

    Combination of ICI with adoptive cellular therapy

    TILs can be utilized to generate immune cells that are less influenced by the suppressive TME. In the context of ICI therapy, T cells may exhibit dysfunction, which can diminish their effectiveness in some patients. Employing TILs has been proposed as a method to address this T cell dysfunction. This approach has demonstrated efficacy in specific cancers, such as metastatic NSCLC that progresses despite ICI treatment [298].

    Combination of ICI with nanomedicine

    Recently, integrating ICIs with nanomaterials has emerged as a promising strategy to improve the efficacy of ICIs. Research indicates that nanoparticles may enhance the effectiveness of ICIs while mitigating their adverse effects. Notable applications include targeted drug delivery to specific cells and the development of tumor vaccines aimed at DCs. Combining nanomaterials with ICIs has the potential to convert ‘cold’ tumors into ‘inflamed’ tumors by improving T-cell activation and infiltration [299, 300].

    Despite advancements in these strategies, challenges persist in selecting the ideal patient population for combination therapies, achieving an optimal balance of synergistic antitumor effects, and elucidating the adverse reactions linked to these treatments.

    Conclusions

    Immunotherapy is the treatment paradigm against cancer, with resistance to it being one of the most important challenges in oncology. This review summarizes the role of ICIs, as well as their resistance mechanisms and various approaches to reverse it. Much of the research investment in oncology is focused on finding strategies to enhance and overcome both primary and acquired resistance to ICIs. Hopefully, these strategies will soon become a reality in routine clinical practice, exponentially increasing the number of potential patients with advanced cancer who are ‘cured’ or long-term survivors.

    Abbreviations

    AMPs:

    antimicrobial peptides

    APCs:

    antigen-presenting cells

    cDCs:

    classical dendritic cells

    CTLA-4:

    cytotoxic T-lymphocyte antigen 4

    CTLs:

    cytotoxic T lymphocytes

    DCs:

    dendritic cells

    ECM:

    extracellular matrix

    Fc:

    fragment crystallizable

    FcγRs:

    Fcγ receptors

    FMT:

    fecal microbiota transplantation

    HLA:

    human leukocyte antigen

    ICIs:

    immune checkpoint inhibitors

    IFN:

    interferon

    Ig:

    immunoglobulin

    IgGs:

    immunoglobulin Gs

    IL:

    interleukin

    JAK1/2:

    Janus kinase 1/2

    LAG-3:

    lymphocyte activation gene-3

    MDSCs:

    myeloid derived suppressor cells

    MHC:

    major histocompatibility complex

    NK:

    natural killer

    NSCLC:

    non-small cell lung cancer

    PD-1:

    programmed cell death protein 1

    PD-L1:

    programmed death ligand 1

    SHP-2:

    Src homology region 2 domain-containing phosphatase-2

    STAT:

    signal transducer and activator of transcription

    TAMs:

    tumor-associated macrophages

    TCR:

    T cell receptor

    TGF-β:

    transforming growth factor beta

    TIGIT:

    T-cell Immunoreceptor with Ig and ITIM domains

    TILs:

    tumor-infiltrating lymphocytes

    TIM-3:

    T-cell immunoglobulin and mucin-domain containing-3

    TME:

    tumor microenvironment

    Tregs:

    regulatory T cells

    β2M:

    β2 microglobulin

    Declarations

    Acknowledgments

    This manuscript is dedicated to all patients and their families. They are the primary motivation behind our research efforts. We also acknowledge that Figure 2 was created with the help of ChatGPT and appreciate its contribution to the development of this work.

    Author contributions

    LCG: Data curation, Visualization, Investigation, Writing—review & editing, Writing—original draft. SCC: Conceptualization, Methodology, Software. MPK: Supervision, Investigation. BCO: Methodology, Writing—review & editing, Validation. VPB: Data curation, Validation, Investigation, Writing—review & editing. All authors read and approved the submitted version.

    Conflicts of interest

    Luis Cabezón-Gutiérrez reports he received payment for presentations of Roche, Astra Zeneca, Brystol Myers Squibb, Merck Serono, Ipsen Pharma, Grunenthal, Kyowa Kirin, Pfizer and Eisai and received support for attending meetings from Roche, Merck, Eli Lilly, Bristol-Myers Squibb and Nutricia. Vilma Pacheco-Barcia reports she received a grant as an award from Merck and FSEOM, payment for presentations of Merck, Eli Lilly, Eisai and Pierre Fabre and received support for attending meetings from Roche, Eli Lilly, Bristol-Myers Squibb, Merck, Amgen, Merck Sharp and Dhome, and Nutricia. Vilma Pacheco-Barcia also reports she participated in an advisory board from advanced accelerator applications, a Novartis company. Sara Custodio-Cabello has received honoraria (outside of this submitted study) from Fresenius, Astellas Pharma, Merck and Abbott and received support for attending meetings from Pierre-Fabre and Amgen. Magda Palka-Kotlowska has received payment for presentations of Pfizer, Devon, Pharmamar, and Esteve and received support for attending meetings from Pfizer and Novartis. Beatriz Chacón-Ovejero have no conflicts of interest to declare. All the authors declare that they have no known competing financial interests or personal relationships that could have appeared to influence the work reported in this paper.

    Ethical approval

    Not applicable.

    Consent to participate

    Not applicable.

    Consent to publication

    Not applicable.

    Availability of data and materials

    Not applicable.

    Funding

    Not applicable.

    Copyright

    © The Author(s) 2025.

    Publisher’s note

    Open Exploration maintains a neutral stance on jurisdictional claims in published institutional affiliations and maps. All opinions expressed in this article are the personal views of the author(s) and do not represent the stance of the editorial team or the publisher.

    References

    Powles T, Eder JP, Fine GD, Braiteh FS, Loriot Y, Cruz C, et al. MPDL3280A (anti-PD-L1) treatment leads to clinical activity in metastatic bladder cancer. Nature. 2014;515:55862. [DOI] [PubMed]
    Larkin J, Chiarion-Sileni V, Gonzalez R, Grob JJ, Cowey CL, Lao CD, et al. Combined Nivolumab and Ipilimumab or Monotherapy in Untreated Melanoma. N Engl J Med. 2015;373:2334. Erratum in: N Engl J Med. 2018;379:2185. [DOI] [PubMed] [PMC]
    Borghaei H, Paz-Ares L, Horn L, Spigel DR, Steins M, Ready NE, et al. Nivolumab versus Docetaxel in Advanced Nonsquamous Non–Small-Cell Lung Cancer. N Engl J Med. 2015;373:162739. [DOI] [PubMed] [PMC]
    Gettinger SN, Horn L, Gandhi L, Spigel DR, Antonia SJ, Rizvi NA, et al. Overall Survival and Long-Term Safety of Nivolumab (Anti–Programmed Death 1 Antibody, BMS-936558, ONO-4538) in Patients With Previously Treated Advanced Non–Small-Cell Lung Cancer. J Clin Oncol. 2015;33:200412. [DOI] [PubMed] [PMC]
    Brahmer JR, Tykodi SS, Chow LQM, Hwu W, Topalian SL, Hwu P, et al. Safety and activity of anti–PD-L1 antibody in patients with advanced cancer. N Engl J Med. 2012;366:245565. [DOI] [PubMed] [PMC]
    Herbst RS, Soria JC, Kowanetz M, Fine GD, Hamid O, Gordon MS, et al. Predictive correlates of response to the anti-PD-L1 antibody MPDL3280A in cancer patients. Nature. 2014;515:5637. [DOI] [PubMed] [PMC]
    Gubin MM, Zhang X, Schuster H, Caron E, Ward JP, Noguchi T, et al. Checkpoint blockade cancer immunotherapy targets tumour-specific mutant antigens. Nature. 2014;515:57781. [DOI] [PubMed] [PMC]
    Sucker A, Zhao F, Real B, Heeke C, Bielefeld N, Maβen S, et al. Genetic evolution of T-cell resistance in the course of melanoma progression. Clin Cancer Res. 2014;20:6593604. [DOI] [PubMed] [PMC]
    Liu C, Peng W, Xu C, Lou Y, Zhang M, Wargo JA, et al. BRAF inhibition increases tumor infiltration by T cells and enhances the antitumor activity of adoptive immunotherapy in mice. Clin Cancer Res. 2013;19:393403. [DOI] [PubMed] [PMC]
    Skoulidis F, Goldberg ME, Greenawalt DM, Hellmann MD, Awad MM, Gainor JF, et al. STK11/LKB1 Mutations and PD-1 Inhibitor Resistance in KRAS-Mutant Lung Adenocarcinoma. Cancer Discov. 2018;8:82235. [DOI] [PubMed] [PMC]
    Garon EB, Rizvi NA, Hui R, Leighl N, Balmanoukian AS, Eder JP, et al.; KEYNOTE-001 Investigators. Pembrolizumab for the treatment of non–small-cell lung cancer. N Engl J Med. 2015;372:201828. [DOI] [PubMed]
    Taube JM, Klein A, Brahmer JR, Xu H, Pan X, Kim JH, et al. Association of PD-1, PD-1 ligands, and other features of the tumor immune microenvironment with response to anti–PD-1 therapy. Clin Cancer Res. 2014;20:506474. [DOI] [PubMed] [PMC]
    Postow MA, Chesney J, Pavlick AC, Robert C, Grossmann K, McDermott D, et al. Nivolumab and ipilimumab versus ipilimumab in untreated melanoma. N Engl J Med. 2015;372:200617. Erratum in: N Engl J Med. 2018;379:2185. [DOI] [PubMed] [PMC]
    Zaretsky JM, Garcia-Diaz A, Shin DS, Escuin-Ordinas H, Hugo W, Hu-Lieskovan S, et al. Mutations Associated with Acquired Resistance to PD-1 Blockade in Melanoma. N Engl J Med. 2016;375:81929. [DOI] [PubMed] [PMC]
    Gettinger S, Choi J, Hastings K, Truini A, Datar I, Sowell R, et al. Impaired HLA Class I Antigen Processing and Presentation as a Mechanism of Acquired Resistance to Immune Checkpoint Inhibitors in Lung Cancer. Cancer Discov. 2017;7:142035. [DOI] [PubMed] [PMC]
    Koyama S, Akbay EA, Li YY, Herter-Sprie GS, Buczkowski KA, Richards WG, et al. Adaptive resistance to therapeutic PD-1 blockade is associated with upregulation of alternative immune checkpoints. Nat Commun. 2016;7:10501. [DOI] [PubMed] [PMC]
    Paulson KG, Voillet V, McAfee MS, Hunter DS, Wagener FD, Perdicchio M, et al. Acquired cancer resistance to combination immunotherapy from transcriptional loss of class I HLA. Nat Commun. 2018;9:3868. [DOI] [PubMed] [PMC]
    Biggs MJP, Milone MC, Santos LC, Gondarenko A, Wind SJ. High-resolution imaging of the immunological synapse and T-cell receptor microclustering through microfabricated substrates. J R Soc Interface. 2011;8:146271. [DOI] [PubMed] [PMC]
    Wherry EJ. T cell exhaustion. Nat Immunol. 2011;12:4929. [DOI] [PubMed]
    Schwartz RH. A cell culture model for T lymphocyte clonal anergy. Science. 1990;248:134956. [DOI] [PubMed]
    Navarro AG, Björklund AT, Chekenya M. Therapeutic potential and challenges of natural killer cells in treatment of solid tumors. Front Immunol. 2015;6:202. [DOI] [PubMed] [PMC]
    Laoui D, Van Overmeire E, De Baetselier P, Van Ginderachter JA, Raes G. Functional Relationship between Tumor-Associated Macrophages and Macrophage Colony-Stimulating Factor as Contributors to Cancer Progression. Front Immunol. 2014;5:489. [DOI] [PubMed] [PMC]
    Savage PA, Leventhal DS, Malchow S. Shaping the repertoire of tumor-infiltrating effector and regulatory T cells. Immunol Rev. 2014;259:24558. [DOI] [PubMed] [PMC]
    Marvel D, Gabrilovich DI. Myeloid-derived suppressor cells in the tumor microenvironment: expect the unexpected. J Clin Invest. 2015;125:335664. [DOI] [PubMed] [PMC]
    van der Merwe PA, Dushek O. Mechanisms for T cell receptor triggering. Nat Rev Immunol. 2011;11:4755. [DOI] [PubMed]
    Aggarwal V, Workman CJ, Vignali DAA. LAG-3 as the third checkpoint inhibitor. Nat Immunol. 2023;24:141522. [DOI] [PubMed] [PMC]
    Tawbi HA, Schadendorf D, Lipson EJ, Ascierto PA, Matamala L, Gutiérrez EC, et al.; RELATIVITY-047 Investigators. Relatlimab and Nivolumab versus Nivolumab in Untreated Advanced Melanoma. N Engl J Med. 2022;386:2434. [DOI] [PubMed] [PMC]
    Bagchi S, Yuan R, Engleman EG. Immune Checkpoint Inhibitors for the Treatment of Cancer: Clinical Impact and Mechanisms of Response and Resistance. Annu Rev Pathol. 2021;16:22349. [DOI] [PubMed]
    Larkin J, Minor D, D’Angelo S, Neyns B, Smylie M, Miller WH Jr, et al. Overall Survival in Patients With Advanced Melanoma Who Received Nivolumab Versus Investigator’s Choice Chemotherapy in CheckMate 037: A Randomized, Controlled, Open-Label Phase III Trial. J Clin Oncol. 2018;36:38390. [DOI] [PubMed] [PMC]
    Weber J, Mandala M, Vecchio MD, Gogas HJ, Arance AM, Cowey CL, et al. Adjuvant Nivolumab versus Ipilimumab in Resected Stage III or IV Melanoma. N Engl J Med. 2017;377:182435. [DOI] [PubMed]
    Forde PM, Spicer J, Lu S, Provencio M, Mitsudomi T, Awad MM, et al.; CheckMate 816 Investigators. Neoadjuvant Nivolumab plus Chemotherapy in Resectable Lung Cancer. N Engl J Med. 2022;386:197385. [DOI] [PubMed] [PMC]
    Borghaei H, Gettinger S, Vokes EE, Chow LQM, Burgio MA, de Castro Carpeno J, et al. Five-Year Outcomes From the Randomized, Phase III Trials CheckMate 017 and 057: Nivolumab Versus Docetaxel in Previously Treated Non–Small-Cell Lung Cancer. J Clin Oncol. 2021;39:72333. Erratum in: J Clin Oncol. 2021;39:1190. [DOI] [PubMed] [PMC]
    Motzer RJ, Escudier B, McDermott DF, George S, Hammers HJ, Srinivas S, et al.; CheckMate 025 Investigators. Nivolumab versus Everolimus in Advanced Renal-Cell Carcinoma. N Engl J Med. 2015;373:180313. [DOI] [PubMed] [PMC]
    Choueiri TK, Powles T, Burotto M, Escudier B, Bourlon MT, Zurawski B, et al.; CheckMate 9ER Investigators. Nivolumab plus Cabozantinib versus Sunitinib for Advanced Renal-Cell Carcinoma. N Engl J Med. 2021;384:82941. [DOI] [PubMed] [PMC]
    Ferris RL, Blumenschein G Jr, Fayette J, Guigay J, Colevas AD, Licitra L, et al. Nivolumab for Recurrent Squamous-Cell Carcinoma of the Head and Neck. N Engl J Med. 2016;375:185667. [DOI] [PubMed] [PMC]
    Bajorin DF, Witjes JA, Gschwend JE, Schenker M, Valderrama BP, Tomita Y, et al. Adjuvant Nivolumab versus Placebo in Muscle-Invasive Urothelial Carcinoma. N Engl J Med. 2021;384:210214. Erratum in: N Engl J Med. 2021;385:864. [DOI] [PubMed] [PMC]
    van der Heijden MS, Sonpavde G, Powles T, Necchi A, Burotto M, Schenker M, et al.; CheckMate 901 Trial Investigators. Nivolumab plus Gemcitabine–Cisplatin in Advanced Urothelial Carcinoma. N Engl J Med. 2023;389:177889. [DOI] [PubMed]
    Sharma P, Retz M, Siefker-Radtke A, Baron A, Necchi A, Bedke J, et al. Nivolumab in metastatic urothelial carcinoma after platinum therapy (CheckMate 275): a multicentre, single-arm, phase 2 trial. Lancet Oncol. 2017;18:31222. [DOI] [PubMed]
    Kelly RJ, Ajani JA, Kuzdzal J, Zander T, Cutsem EV, Piessen G, et al.; CheckMate 577 Investigators. Adjuvant Nivolumab in Resected Esophageal or Gastroesophageal Junction Cancer. N Engl J Med. 2021;384:1191203. Erratum in: N Engl J Med. 2023;388:672. [DOI] [PubMed]
    Doki Y, Ajani JA, Kato K, Xu J, Wyrwicz L, Motoyama S, et al.; CheckMate 648 Trial Investigators. Nivolumab Combination Therapy in Advanced Esophageal Squamous-Cell Carcinoma. N Engl J Med. 2022;386:44962. [DOI] [PubMed]
    Kato K, Cho BC, Takahashi M, Okada M, Lin C, Chin K, et al. Nivolumab versus chemotherapy in patients with advanced oesophageal squamous cell carcinoma refractory or intolerant to previous chemotherapy (ATTRACTION-3): a multicentre, randomised, open-label, phase 3 trial. Lancet Oncol. 2019;20:150617. Erratum in: Lancet Oncol. 2019;20:e613. [DOI] [PubMed]
    Janjigian YY, Shitara K, Moehler M, Garrido M, Salman P, Shen L, et al. First-line nivolumab plus chemotherapy versus chemotherapy alone for advanced gastric, gastro-oesophageal junction, and oesophageal adenocarcinoma (CheckMate 649): a randomised, open-label, phase 3 trial. Lancet. 2021;398:2740. [DOI] [PubMed] [PMC]
    Luke JJ, Rutkowski P, Queirolo P, Vecchio MD, Mackiewicz J, Chiarion-Sileni V, et al.; KEYNOTE-716 Investigators. Pembrolizumab versus placebo as adjuvant therapy in completely resected stage IIB or IIC melanoma (KEYNOTE-716): a randomised, double-blind, phase 3 trial. Lancet. 2022;399:171829. [DOI] [PubMed]
    Schachter J, Ribas A, Long GV, Arance A, Grob J, Mortier L, et al. Pembrolizumab versus ipilimumab for advanced melanoma: final overall survival results of a multicentre, randomised, open-label phase 3 study (KEYNOTE-006). Lancet. 2017;390:185362. [DOI] [PubMed]
    Reck M, Rodríguez-Abreu D, Robinson AG, Hui R, Csőszi T, Fülöp A, et al. Updated Analysis of KEYNOTE-024: Pembrolizumab Versus Platinum-Based Chemotherapy for Advanced Non–Small-Cell Lung Cancer With PD-L1 Tumor Proportion Score of 50% or Greater. J Clin Oncol. 2019;37:53746. [DOI] [PubMed]
    Gandhi L, Rodríguez-Abreu D, Gadgeel S, Esteban E, Felip E, Angelis FD, et al.; KEYNOTE-189 Investigators. Pembrolizumab plus Chemotherapy in Metastatic Non–Small-Cell Lung Cancer. N Engl J Med. 2018;378:207892. [DOI] [PubMed]
    Paz-Ares L, Luft A, Vicente D, Tafreshi A, Gümüş M, Mazières J, et al.; KEYNOTE-407 Investigators. Pembrolizumab plus Chemotherapy for Squamous Non–Small-Cell Lung Cancer. N Engl J Med. 2018;379:204051. [DOI] [PubMed]
    Herbst RS, Baas P, Kim D, Felip E, Pérez-Gracia JL, Han J, et al. Pembrolizumab versus docetaxel for previously treated, PD-L1-positive, advanced non-small-cell lung cancer (KEYNOTE-010): a randomised controlled trial. Lancet. 2016;387:154050. [DOI] [PubMed]
    Bellmunt J, de Wit R, Vaughn DJ, Fradet Y, Lee J, Fong L, et al.; KEYNOTE-045 Investigators. Pembrolizumab as Second-Line Therapy for Advanced Urothelial Carcinoma. N Engl J Med. 2017;376:101526. [DOI] [PubMed] [PMC]
    Balar AV, Castellano D, O'Donnell PH, Grivas P, Vuky J, Powles T, et al. First-line pembrolizumab in cisplatin-ineligible patients with locally advanced and unresectable or metastatic urothelial cancer (KEYNOTE-052): a multicentre, single-arm, phase 2 study. Lancet Oncol. 2017;18:148392. [DOI] [PubMed]
    Burtness B, Harrington KJ, Greil R, Soulières D, Tahara M, de Castro G Jr, et al.; KEYNOTE-048 Investigators. Pembrolizumab alone or with chemotherapy versus cetuximab with chemotherapy for recurrent or metastatic squamous cell carcinoma of the head and neck (KEYNOTE-048): a randomised, open-label, phase 3 study. Lancet. 2019;394:191528. [DOI] [PubMed]
    Choueiri TK, Tomczak P, Park SH, Venugopal B, Ferguson T, Chang Y, et al.; KEYNOTE-564 Investigators. Adjuvant Pembrolizumab after Nephrectomy in Renal-Cell Carcinoma. N Engl J Med. 2021;385:68394. [DOI] [PubMed]
    Rini BI, Plimack ER, Stus V, Gafanov R, Hawkins R, Nosov D, et al.; KEYNOTE-426 Investigators. Pembrolizumab plus Axitinib versus Sunitinib for Advanced Renal-Cell Carcinoma. N Engl J Med. 2019;380:111627. [DOI] [PubMed]
    Motzer R, Alekseev B, Rha SY, Porta C, Eto M, Powles T, et al.; CLEAR Trial Investigators. Lenvatinib plus Pembrolizumab or Everolimus for Advanced Renal Cell Carcinoma. N Engl J Med. 2021;384:1289300. [DOI] [PubMed]
    André T, Shiu K, Kim TW, Jensen BV, Jensen LH, Punt C, et al.; KEYNOTE-177 Investigators. Pembrolizumab in Microsatellite-Instability–High Advanced Colorectal Cancer. N Engl J Med. 2020;383:220718. [DOI] [PubMed]
    Marabelle A, Le DT, Ascierto PA, Di Giacomo AM, De Jesus-Acosta A, Delord JP, et al. Efficacy of Pembrolizumab in Patients With Noncolorectal High Microsatellite Instability/Mismatch Repair-Deficient Cancer: Results From the Phase II KEYNOTE-158 Study. J Clin Oncol. 2020;38:110. [DOI] [PubMed] [PMC]
    Sun J, Shen L, Shah MA, Enzinger P, Adenis A, Doi T, et al.; KEYNOTE-590 Investigators. Pembrolizumab plus chemotherapy versus chemotherapy alone for first-line treatment of advanced oesophageal cancer (KEYNOTE-590): a randomised, placebo-controlled, phase 3 study. Lancet. 2021;398:75971. Erratum in: Lancet. 2021;398:1874. [DOI] [PubMed]
    Schmid P, Cortes J, Pusztai L, McArthur H, Kümmel S, Bergh J, et al.; KEYNOTE-522 Investigators. Pembrolizumab for Early Triple-Negative Breast Cancer. N Engl J Med. 2020;382:81021. [DOI] [PubMed]
    Cortes J, Cescon DW, Rugo HS, Nowecki Z, Im S, Yusof MM, et al.; KEYNOTE-355 Investigators. Pembrolizumab plus chemotherapy versus placebo plus chemotherapy for previously untreated locally recurrent inoperable or metastatic triple-negative breast cancer (KEYNOTE-355): a randomised, placebo-controlled, double-blind, phase 3 clinical trial. Lancet. 2020;396:181728. [DOI] [PubMed]
    Makker V, Colombo N, Herráez AC, Santin AD, Colomba E, Miller DS, et al.; Study 309–KEYNOTE-775 Investigators. Lenvatinib plus Pembrolizumab for Advanced Endometrial Cancer. N Engl J Med. 2022;386:43748. [DOI] [PubMed] [PMC]
    Monk BJ, Colombo N, Tewari KS, Dubot C, Caceres MV, Hasegawa K, et al.; KEYNOTE-826 Investigators. First-Line Pembrolizumab + Chemotherapy Versus Placebo + Chemotherapy for Persistent, Recurrent, or Metastatic Cervical Cancer: Final Overall Survival Results of KEYNOTE-826. J Clin Oncol. 2023;41:550511. [DOI] [PubMed]
    Janjigian YY, Kawazoe A, Bai Y, Xu J, Lonardi S, Metges JP, et al.; KEYNOTE-811 Investigators. Pembrolizumab plus trastuzumab and chemotherapy for HER2-positive gastric or gastro-oesophageal junction adenocarcinoma: interim analyses from the phase 3 KEYNOTE-811 randomised placebo-controlled trial. Lancet. 2023;402:2197208. [DOI] [PubMed]
    Rha SY, Oh D, Yañez P, Bai Y, Ryu M, Lee J, et al. Pembrolizumab plus chemotherapy versus placebo plus chemotherapy for HER2-negative advanced gastric cancer (KEYNOTE-859): a multicentre, randomised, double-blind, phase 3 trial. Lancet Oncol. 2023;24:118195. Erratum in: Lancet Oncol. 2024;25:e626. [DOI] [PubMed]
    Kelley RK, Ueno M, Yoo C, Finn RS, Furuse J, Ren Z, et al.; KEYNOTE-966 Investigators. Pembrolizumab in combination with gemcitabine and cisplatin compared with gemcitabine and cisplatin alone for patients with advanced biliary tract cancer (KEYNOTE-966): a randomised, double-blind, placebo-controlled, phase 3 trial. Lancet. 2023;401:185365. Erratum in: Lancet. 2023;402:964. Erratum in: Lancet. 2024;403:1140. [DOI] [PubMed]
    Mirza MR, Chase DM, Slomovitz BM, dePont Christensen R, Novák Z, Black D, et al.; RUBY Investigators. Dostarlimab for Primary Advanced or Recurrent Endometrial Cancer. N Engl J Med. 2023;388:214558. [DOI] [PubMed]
    Oaknin A, Gilbert L, Tinker AV, Brown J, Mathews C, Press J, et al. Safety and antitumor activity of dostarlimab in patients with advanced or recurrent DNA mismatch repair deficient/microsatellite instability-high (dMMR/MSI-H) or proficient/stable (MMRp/MSS) endometrial cancer: interim results from GARNET—a phase I, single-arm study. J Immunother Cancer. 2022;10:e003777. [DOI] [PubMed] [PMC]
    Migden MR, Rischin D, Schmults CD, Guminski A, Hauschild A, Lewis KD, et al. PD-1 Blockade with Cemiplimab in Advanced Cutaneous Squamous-Cell Carcinoma. N Engl J Med. 2018;379:34151. [DOI] [PubMed]
    Stratigos AJ, Sekulic A, Peris K, Bechter O, Prey S, Kaatz M, et al. Cemiplimab in locally advanced basal cell carcinoma after hedgehog inhibitor therapy: an open-label, multi-centre, single-arm, phase 2 trial. Lancet Oncol. 2021;22:84857. [DOI] [PubMed]
    Sezer A, Kilickap S, Gümüş M, Bondarenko I, Özgüroğlu M, Gogishvili M, et al. Cemiplimab monotherapy for first-line treatment of advanced non-small-cell lung cancer with PD-L1 of at least 50%: a multicentre, open-label, global, phase 3, randomised, controlled trial. Lancet. 2021;397:592604. [DOI] [PubMed]
    Gogishvili M, Melkadze T, Makharadze T, Giorgadze D, Dvorkin M, Penkov K, et al. Cemiplimab plus chemotherapy versus chemotherapy alone in non-small cell lung cancer: a randomized, controlled, double-blind phase 3 trial. Nat Med. 2022;28:237480. [DOI] [PubMed] [PMC]
    Tewari KS, Monk BJ, Vergote I, Miller A, de Melo AC, Kim HS, et al.; Investigators for GOG Protocol 3016 and ENGOT Protocol En-Cx9. Survival with Cemiplimab in Recurrent Cervical Cancer. N Engl J Med. 2022;386:54455. [DOI] [PubMed]
    Lakhani N, Cosman R, Banerji U, Rasco D, Tomaszewska-Kiecana M, Garralda E, et al. A first-in-human phase I study of the PD-1 inhibitor, retifanlimab (INCMGA00012), in patients with advanced solid tumors (POD1UM-101). ESMO Open. 2024;9:102254. [DOI] [PubMed] [PMC]
    Shen L, Kato K, Kim S, Ajani JA, Zhao K, He Z, et al.; RATIONALE-302 Investigators. Tislelizumab Versus Chemotherapy as Second-Line Treatment for Advanced or Metastatic Esophageal Squamous Cell Carcinoma (RATIONALE-302): A Randomized Phase III Study. J Clin Oncol. 2022;40:306576. Erratum in: J Clin Oncol. 2024;42:486. [DOI] [PubMed] [PMC]
    Powles T, Durán I, van der Heijden MS, Loriot Y, Vogelzang NJ, De Giorgi U, et al. Atezolizumab versus chemotherapy in patients with platinum-treated locally advanced or metastatic urothelial carcinoma (IMvigor211): a multicentre, open-label, phase 3 randomised controlled trial. Lancet. 2018;391:74857. Erratum in: Lancet. 2018;392:1402. [DOI] [PubMed]
    Rittmeyer A, Barlesi F, Waterkamp D, Park K, Ciardiello F, von Pawel J, et al.; OAK Study Group. Atezolizumab versus docetaxel in patients with previously treated non-small-cell lung cancer (OAK): a phase 3, open-label, multicentre randomised controlled trial. Lancet. 2017;389:25565. Erratum in: Lancet. 2017;389:e5. [DOI] [PubMed] [PMC]
    D’Angelo SP, Bhatia S, Brohl AS, Hamid O, Mehnert JM, Terheyden P, et al. Avelumab in patients with previously treated metastatic Merkel cell carcinoma: long-term data and biomarker analyses from the single-arm phase 2 JAVELIN Merkel 200 trial. J Immunother Cancer. 2020;8:e000674. [DOI] [PubMed] [PMC]
    Powles T, Park SH, Voog E, Caserta C, Valderrama BP, Gurney H, et al. Avelumab Maintenance Therapy for Advanced or Metastatic Urothelial Carcinoma. N Engl J Med. 2020;383:121830. [DOI] [PubMed]
    Motzer RJ, Penkov K, Haanen J, Rini B, Albiges L, Campbell MT, et al. Avelumab plus Axitinib versus Sunitinib for Advanced Renal-Cell Carcinoma. N Engl J Med. 2019;380:110315. [DOI] [PubMed] [PMC]
    Antonia SJ, Villegas A, Daniel D, Vicente D, Murakami S, Hui R, et al.; PACIFIC Investigators. Durvalumab after Chemoradiotherapy in Stage III Non–Small-Cell Lung Cancer. N Engl J Med. 2017;377:191929. [DOI] [PubMed]
    Paz-Ares L, Dvorkin M, Chen Y, Reinmuth N, Hotta K, Trukhin D, et al. Durvalumab plus platinum-etoposide versus platinum-etoposide in first-line treatment of extensive-stage small-cell lung cancer (CASPIAN): a randomised, controlled, open-label, phase 3 trial. Lancet. 2019;394:192939. [DOI] [PubMed]
    Burris HA 3rd, Okusaka T, Vogel A, Lee MA, Takahashi H, Breder V, et al. Durvalumab plus gemcitabine and cisplatin in advanced biliary tract cancer (TOPAZ-1): patient-reported outcomes from a randomised, double-blind, placebo-controlled, phase 3 trial. Lancet Oncol. 2024;25:62635. [DOI] [PubMed]
    Abou-Alfa GK, Lau G, Kudo M, Chan SL, Kelley RK, Furuse J, et al. Tremelimumab plus Durvalumab in Unresectable Hepatocellular Carcinoma. NEJM Evid. 2022;1:EVIDoa2100070. [DOI] [PubMed]
    Hodi FS, O’Day SJ, McDermott DF, Weber RW, Sosman JA, Haanen JB, et al. Improved survival with ipilimumab in patients with metastatic melanoma. N Engl J Med. 2010;363:71123. Erratum in: N Engl J Med. 2010;363:1290. [DOI] [PubMed] [PMC]
    Motzer RJ, Tannir NM, McDermott DF, Frontera OA, Melichar B, Choueiri TK, et al.; CheckMate 214 Investigators. Nivolumab plus Ipilimumab versus Sunitinib in Advanced Renal-Cell Carcinoma. N Engl J Med. 2018;378:127790. [DOI] [PubMed] [PMC]
    Baas P, Scherpereel A, Nowak AK, Fujimoto N, Peters S, Tsao AS, et al. First-line nivolumab plus ipilimumab in unresectable malignant pleural mesothelioma (CheckMate 743): a multicentre, randomised, open-label, phase 3 trial. Lancet. 2021;397:37586. [DOI] [PubMed]
    Kato K, Doki Y, Chau I, Xu J, Wyrwicz L, Motoyama S, et al. Nivolumab plus chemotherapy or ipilimumab versus chemotherapy in patients with advanced esophageal squamous cell carcinoma (CheckMate 648): 29-month follow-up from a randomized, open-label, phase III trial. Cancer Med. 2024;13:e7235. Erratum in: Cancer Med. 2025;14:e70652. [DOI] [PubMed] [PMC]
    Lenz HJ, Van Cutsem E, Luisa Limon M, Wong KYM, Hendlisz A, Aglietta M, et al. First-Line Nivolumab Plus Low-Dose Ipilimumab for Microsatellite Instability-High/Mismatch Repair-Deficient Metastatic Colorectal Cancer: The Phase II CheckMate 142 Study. J Clin Oncol. 2022;40:16170. [DOI] [PubMed]
    Johnson ML, Cho BC, Luft A, Alatorre-Alexander J, Geater SL, Laktionov K, et al. Durvalumab With or Without Tremelimumab in Combination With Chemotherapy as First-Line Therapy for Metastatic Non–Small-Cell Lung Cancer: The Phase III POSEIDON Study. J Clin Oncol. 2023;41:121327. [DOI] [PubMed] [PMC]
    Arce Vargas F, Furness AJS, Litchfield K, Joshi K, Rosenthal R, Ghorani E, et al.; TRACERx Melanoma; TRACERx Renal; TRACERx Lung consortia; Pule M, Marafioti T, Gore M, Larkin J, Turajlic S, Swanton C, et al. Fc Effector Function Contributes to the Activity of Human Anti-CTLA-4 Antibodies. Cancer Cell. 2018;33:64963.e4. [DOI] [PubMed] [PMC]
    Linsley PS, Clark EA, Ledbetter JA. T-cell antigen CD28 mediates adhesion with B cells by interacting with activation antigen B7/BB-1. Proc Natl Acad Sci U S A. 1990;87:50315. [DOI] [PubMed] [PMC]
    Linsley PS, Brady W, Grosmaire L, Aruffo A, Damle NK, Ledbetter JA. Binding of the B cell activation antigen B7 to CD28 costimulates T cell proliferation and interleukin 2 mRNA accumulation. J Exp Med. 1991;173:72130. [DOI] [PubMed] [PMC]
    Azuma M, Ito D, Yagita H, Okumura K, Phillips JH, Lanier LL, et al. B70 antigen is a second ligand for CTLA-4 and CD28. Nature. 1993;366:769. [DOI] [PubMed]
    Valk E, Rudd CE, Schneider H. CTLA-4 trafficking and surface expression. Trends Immunol. 2008;29:2729. [DOI] [PubMed] [PMC]
    Walunas TL, Lenschow DJ, Bakker CY, Linsley PS, Freeman GJ, Green JM, et al. CTLA-4 can function as a negative regulator of T cell activation. Immunity. 1994;1:40513. [DOI] [PubMed]
    Krummel MF, Allison JP. CD28 and CTLA-4 have opposing effects on the response of T cells to stimulation. J Exp Med. 1995;182:45965. [DOI] [PubMed] [PMC]
    Valk E, Leung R, Kang H, Kaneko K, Rudd CE, Schneider H. T cell receptor-interacting molecule acts as a chaperone to modulate surface expression of the CTLA-4 coreceptor. Immunity. 2006;25:80721. [DOI] [PubMed]
    Linsley PS, Brady W, Urnes M, Grosmaire LS, Damle NK, Ledbetter JA. CTLA-4 is a second receptor for the B cell activation antigen B7. J Exp Med. 1991;174:5619. [DOI] [PubMed] [PMC]
    Masteller EL, Chuang E, Mullen AC, Reiner SL, Thompson CB. Structural analysis of CTLA-4 function in vivo. J Immunol. 2000;164:531927. [DOI] [PubMed]
    Chuang E, Fisher TS, Morgan RW, Robbins MD, Duerr JM, Heiden MGV, et al. The CD28 and CTLA-4 receptors associate with the serine/threonine phosphatase PP2A. Immunity. 2000;13:31322. [DOI] [PubMed]
    Chuang E, Lee KM, Robbins MD, Duerr JM, Alegre ML, Hambor JE, et al. Regulation of cytotoxic T lymphocyte-associated molecule-4 by Src kinases. J Immunol. 1999;162:12707. [PubMed]
    Lee KM, Chuang E, Griffin M, Khattri R, Hong DK, Zhang W, et al. Molecular basis of T cell inactivation by CTLA-4. Science. 1998;282:22636. [DOI] [PubMed]
    Marengère LE, Waterhouse P, Duncan GS, Mittrücker HW, Feng GS, Mak TW. Regulation of T cell receptor signaling by tyrosine phosphatase SYP association with CTLA-4. Science. 1996;272:11703. Erratum in: Science 1996;274:1597. Erratum in: Science 1997;276:21. [DOI] [PubMed]
    Fraser JH, Rincón M, McCoy KD, Gros GL. CTLA4 ligation attenuates AP-1, NFAT and NF-κB activity in activated T cells. Eur J Immunol. 1999;29:83844. [DOI] [PubMed]
    Olsson C, Riesbeck K, Dohlsten M, Michaëlsson E. CTLA-4 ligation suppresses CD28-induced NF-κB and AP-1 activity in mouse T cell blasts. J Biol Chem. 1999;274:144005. Erratum in: J Biol Chem. 1999;274:21490. [DOI] [PubMed]
    Hoff H, Kolar P, Ambach A, Radbruch A, Brunner-Weinzierl MC. CTLA-4 (CD152) inhibits T cell function by activating the ubiquitin ligase Itch. Mol Immunol. 2010;47:187581. [DOI] [PubMed]
    Read S, Malmström V, Powrie F. Cytotoxic T lymphocyte–associated antigen 4 plays an essential role in the function of Cd25+Cd4+ regulatory cells that control intestinal inflammation. J Exp Med. 2000;192:295302. [DOI] [PubMed] [PMC]
    Takahashi T, Tagami T, Yamazaki S, Uede T, Shimizu J, Sakaguchi N, et al. Immunologic self-tolerance maintained by Cd25+Cd4+ regulatory T cells constitutively expressing cytotoxic T lymphocyte–associated antigen 4. J Exp Med. 2000;192:30310. [DOI] [PubMed] [PMC]
    Wing K, Onishi Y, Prieto-Martin P, Yamaguchi T, Miyara M, Fehervari Z, et al. CTLA-4 control over Foxp3+ regulatory T cell function. Science. 2008;322:2715. [DOI] [PubMed]
    Friedline RH, Brown DS, Nguyen H, Kornfeld H, Lee J, Zhang Y, et al. CD4+ regulatory T cells require CTLA-4 for the maintenance of systemic tolerance. J Exp Med. 2009;206:42134. Erratum in: J Exp Med. 2009;206:721. [DOI] [PubMed] [PMC]
    Zheng SG, Wang JH, Stohl W, Kim KS, Gray JD, Horwitz DA. TGF-β requires CTLA-4 early after T cell activation to induce FoxP3 and generate adaptive CD4+CD25+ regulatory cells. J Immunol. 2006;176:33219. [DOI] [PubMed]
    Sharma A, Subudhi SK, Blando J, Scutti J, Vence L, Wargo J, et al. Anti-CTLA-4 Immunotherapy Does Not Deplete FOXP3+ Regulatory T Cells (Tregs) in Human Cancers. Clin Cancer Res. 2019;25:12338. [DOI] [PubMed] [PMC]
    Lax BM, Palmeri JR, Lutz EA, Sheen A, Stinson JA, Duhamel L, et al. Both intratumoral regulatory T cell depletion and CTLA-4 antagonism are required for maximum efficacy of anti-CTLA-4 antibodies. Proc Natl Acad Sci U S A. 2023;120:e2300895120. [DOI] [PubMed] [PMC]
    Leach DR, Krummel MF, Allison JP. Enhancement of antitumor immunity by CTLA-4 blockade. Science. 1996;271:17346. [DOI] [PubMed]
    Okazaki T, Maeda A, Nishimura H, Kurosaki T, Honjo T. PD-1 immunoreceptor inhibits B cell receptor-mediated signaling by recruiting src homology 2-domain-containing tyrosine phosphatase 2 to phosphotyrosine. Proc Natl Acad Sci U S A. 2001;98:1386671. [DOI] [PubMed] [PMC]
    Ishida Y, Agata Y, Shibahara K, Honjo T. Induced expression of PD-1, a novel member of the immunoglobulin gene superfamily, upon programmed cell death. EMBO J. 1992;11:388795. [DOI] [PubMed] [PMC]
    Ahmadzadeh M, Johnson LA, Heemskerk B, Wunderlich JR, Dudley ME, White DE, et al. Tumor antigen-specific CD8 T cells infiltrating the tumor express high levels of PD-1 and are functionally impaired. Blood. 2009;114:153744. [DOI] [PubMed] [PMC]
    Salmaninejad A, Valilou SF, Shabgah AG, Aslani S, Alimardani M, Pasdar A, et al. PD-1/PD-L1 pathway: Basic biology and role in cancer immunotherapy. J Cell Physiol. 2019;234:1682437. [DOI] [PubMed]
    Freeman GJ, Long AJ, Iwai Y, Bourque K, Chernova T, Nishimura H, et al. Engagement of the Pd-1 immunoinhibitory receptor by a novel B7 family member leads to negative regulation of lymphocyte activation. J Exp Med. 2000;192:102734. [DOI] [PubMed] [PMC]
    Latchman Y, Wood CR, Chernova T, Chaudhary D, Borde M, Chernova I, et al. PD-L2 is a second ligand for PD-1 and inhibits T cell activation. Nat Immunol. 2001;2:2618. [DOI] [PubMed]
    Anagnostou V, Smith KN, Forde PM, Niknafs N, Bhattacharya R, White J, et al. Evolution of Neoantigen Landscape during Immune Checkpoint Blockade in Non-Small Cell Lung Cancer. Cancer Discov. 2017;7:26476. [DOI] [PubMed] [PMC]
    Sheppard K, Fitz LJ, Lee JM, Benander C, George JA, Wooters J, et al. PD-1 inhibits T-cell receptor induced phosphorylation of the ZAP70/CD3ζ signalosome and downstream signaling to PKCθ. FEBS Lett. 2004;574:3741. [DOI] [PubMed]
    Patsoukis N, Brown J, Petkova V, Liu F, Li L, Boussiotis VA. Selective effects of PD-1 on Akt and Ras pathways regulate molecular components of the cell cycle and inhibit T cell proliferation. Sci Signal. 2012;5:ra46. [DOI] [PubMed] [PMC]
    Nurieva R, Thomas S, Nguyen T, Martin-Orozco N, Wang Y, Kaja M, et al. T-cell tolerance or function is determined by combinatorial costimulatory signals. EMBO J. 2006;25:262333. [DOI] [PubMed] [PMC]
    Chocarro L, Blanco E, Fernandez-Rubio L, Garnica M, Zuazo M, Garcia MJ, et al. PD-1/LAG-3 co-signaling profiling uncovers CBL ubiquitin ligases as key immunotherapy targets. EMBO Mol Med. 2024;16:1791816. [DOI] [PubMed] [PMC]
    Iwai Y, Ishida M, Tanaka Y, Okazaki T, Honjo T, Minato N. Involvement of PD-L1 on tumor cells in the escape from host immune system and tumor immunotherapy by PD-L1 blockade. Proc Natl Acad Sci U S A. 2002;99:122937. [DOI] [PubMed] [PMC]
    Hirano F, Kaneko K, Tamura H, Dong H, Wang S, Ichikawa M, et al. Blockade of B7-H1 and PD-1 by monoclonal antibodies potentiates cancer therapeutic immunity. Cancer Res. 2005;65:108996. [PubMed]
    Curiel TJ, Wei S, Dong H, Alvarez X, Cheng P, Mottram P, et al. Blockade of B7-H1 improves myeloid dendritic cell-mediated antitumor immunity. Nat Med. 2003;9:5627. [DOI] [PubMed]
    He Y, Zhang G, Wang X, Zhang H, Yuan Y, Li D, et al. Blocking programmed death-1 ligand-PD-1 interactions by local gene therapy results in enhancement of antitumor effect of secondary lymphoid tissue chemokine. J Immunol. 2004;173:491928. [DOI] [PubMed]
    Blank C, Brown I, Peterson AC, Spiotto M, Iwai Y, Honjo T, et al. PD-L1/B7H-1 inhibits the effector phase of tumor rejection by T cell receptor (TCR) transgenic CD8+ T cells. Cancer Res. 2004;64:11405. [DOI] [PubMed]
    Iwai Y, Terawaki S, Honjo T. PD-1 blockade inhibits hematogenous spread of poorly immunogenic tumor cells by enhanced recruitment of effector T cells. Int Immunol. 2005;17:13344. [DOI] [PubMed]
    Thompson RH, Gillett MD, Cheville JC, Lohse CM, Dong H, Webster WS, et al. Costimulatory B7-H1 in renal cell carcinoma patients: Indicator of tumor aggressiveness and potential therapeutic target. Proc Natl Acad Sci U S A. 2004;101:171749. [DOI] [PubMed] [PMC]
    Thompson RH, Kuntz SM, Leibovich BC, Dong H, Lohse CM, Webster WS, et al. Tumor B7-H1 is associated with poor prognosis in renal cell carcinoma patients with long-term follow-up. Cancer Res. 2006;66:33815. [DOI] [PubMed]
    Ohigashi Y, Sho M, Yamada Y, Tsurui Y, Hamada K, Ikeda N, et al. Clinical significance of programmed death-1 ligand-1 and programmed death-1 ligand-2 expression in human esophageal cancer. Clin Cancer Res. 2005;11:294753. [DOI] [PubMed]
    Hamanishi J, Mandai M, Iwasaki M, Okazaki T, Tanaka Y, Yamaguchi K, et al. Programmed cell death 1 ligand 1 and tumor-infiltrating CD8+ T lymphocytes are prognostic factors of human ovarian cancer. Proc Natl Acad Sci U S A. 2007;104:33605. [DOI] [PubMed] [PMC]
    Thompson RH, Dong H, Lohse CM, Leibovich BC, Blute ML, Cheville JC, et al. PD-1 is expressed by tumor-infiltrating immune cells and is associated with poor outcome for patients with renal cell carcinoma. Clin Cancer Res. 2007;13:175761. [DOI] [PubMed]
    Mayoux M, Roller A, Pulko V, Sammicheli S, Chen S, Sum E, et al. Dendritic cells dictate responses to PD-L1 blockade cancer immunotherapy. Sci Transl Med. 2020;12:eaav7431. Erratum in: Sci Transl Med. 2020;12:eabd0088. [DOI] [PubMed]
    Curigliano G, Gelderblom H, Mach N, Doi T, Tai WMD, Forde P, et al. Abstract CT183: Phase (Ph) I/II study of MBG453± spartalizumab (PDR001) in patients (pts) with advanced malignancies. Cancer Res. 2019;79:CT183. [DOI]
    Falchook GS, Ribas A, Davar D, Eroglu Z, Wang JS, Luke JJ, et al. Phase 1 trial of TIM-3 inhibitor cobolimab monotherapy and in combination with PD-1 inhibitors nivolumab or dostarlimab (AMBER). J Clin Oncol. 2022;40:2504. [DOI]
    Niu J, Maurice-Dror C, Lee DH, Kim D, Nagrial A, Voskoboynik M, et al. First-in-human phase 1 study of the anti-TIGIT antibody vibostolimab as monotherapy or with pembrolizumab for advanced solid tumors, including non-small-cell lung cancer. Ann Oncol. 2022;33:16980. [DOI] [PubMed]
    Felip E, Majem M, Doger B, Clay TD, Carcereny E, Bondarenko I, et al. A phase II study (TACTI-002) in first-line metastatic non–small cell lung carcinoma investigating eftilagimod alpha (soluble LAG-3 protein) and pembrolizumab: updated results from a PD-L1 unselected population. J Clin Oncol. 2022;40:9003. [DOI]
    Sharpe AH, Wherry EJ, Ahmed R, Freeman GJ. The function of programmed cell death 1 and its ligands in regulating autoimmunity and infection. Nat Immunol. 2007;8:23945. [DOI] [PubMed]
    Butte MJ, Keir ME, Phamduy TB, Sharpe AH, Freeman GJ. Programmed death-1 ligand 1 interacts specifically with the B7-1 costimulatory molecule to inhibit T cell responses. Immunity. 2007;27:11122. [DOI] [PubMed] [PMC]
    Karwacz K, Bricogne C, MacDonald D, Arce F, Bennett CL, Collins M, et al. PD-L1 co-stimulation contributes to ligand-induced T cell receptor down-modulation on CD8+ T cells. EMBO Mol Med. 2011;3:58192. [DOI] [PubMed] [PMC]
    Garcia-Diaz A, Shin DS, Moreno BH, Saco J, Escuin-Ordinas H, Rodriguez GA, et al. Interferon Receptor Signaling Pathways Regulating PD-L1 and PD-L2 Expression. Cell Rep. 2017;19:1189201. Erratum in: Cell Rep. 2019;29:3766. [DOI] [PubMed] [PMC]
    Pardoll DM. The blockade of immune checkpoints in cancer immunotherapy. Nat Rev Cancer. 2012;12:25264. [DOI] [PubMed] [PMC]
    Ribas A. Adaptive Immune Resistance: How Cancer Protects from Immune Attack. Cancer Discov. 2015;5:9159. [DOI] [PubMed] [PMC]
    Wang Y, Du J, Gao Z, Sun H, Mei M, Wang Y, et al. Evolving landscape of PD-L2: bring new light to checkpoint immunotherapy. Br J Cancer. 2023;128:1196207. [DOI] [PubMed] [PMC]
    Kisielow M, Kisielow J, Capoferri-Sollami G, Karjalainen K. Expression of lymphocyte activation gene 3 (LAG-3) on B cells is induced by T cells. Eur J Immunol. 2005;35:20818. [DOI] [PubMed]
    Triebel F, Jitsukawa S, Baixeras E, Roman-Roman S, Genevee C, Viegas-Pequignot E, et al. LAG-3, a novel lymphocyte activation gene closely related to CD4. J Exp Med. 1990;171:1393405. [DOI] [PubMed] [PMC]
    Grosso JF, Goldberg MV, Getnet D, Bruno TC, Yen H, Pyle KJ, et al. Functionally distinct LAG-3 and PD-1 subsets on activated and chronically stimulated CD8 T cells. J Immunol. 2009;182:665969. [DOI] [PubMed] [PMC]
    Workman CJ, Cauley LS, Kim I, Blackman MA, Woodland DL, Vignali DAA. Lymphocyte activation gene-3 (CD223) regulates the size of the expanding T cell population following antigen activation in vivo. J Immunol. 2004;172:54505. [DOI] [PubMed]
    Huang C, Workman CJ, Flies D, Pan X, Marson AL, Zhou G, et al. Role of LAG-3 in regulatory T cells. Immunity. 2004;21:50313. [DOI] [PubMed]
    Workman CJ, Rice DS, Dugger KJ, Kurschner C, Vignali DAA. Phenotypic analysis of the murine CD4-related glycoprotein, CD223 (LAG-3). Eur J Immunol. 2002;32:225563. [DOI] [PubMed]
    Zuazo M, Arasanz H, Fernández-Hinojal G, García-Granda MJ, Gato M, Bocanegra A, et al. Functional systemic CD4 immunity is required for clinical responses to PD-L1/PD-1 blockade therapy. EMBO Mol Med. 2019;11:e10293. [DOI] [PubMed] [PMC]
    Cillo AR, Cardello C, Shan F, Karapetyan L, Kunning S, Sander C, et al. Blockade of LAG-3 and PD-1 leads to co-expression of cytotoxic and exhaustion gene modules in CD8+ T cells to promote antitumor immunity. Cell. 2024;187:437388.e15. [DOI] [PubMed]
    He Y, Yu H, Rozeboom L, Rivard CJ, Ellison K, Dziadziuszko R, et al. LAG-3 Protein Expression in Non–Small Cell Lung Cancer and Its Relationship with PD-1/PD-L1 and Tumor-Infiltrating Lymphocytes. J Thorac Oncol. 2017;12:81423. [DOI] [PubMed]
    Huang R, Francois A, McGray AR, Miliotto A, Odunsi K. Compensatory upregulation of PD-1, LAG-3, and CTLA-4 limits the efficacy of single-agent checkpoint blockade in metastatic ovarian cancer. Oncoimmunology. 2016;6:e1249561. [DOI] [PubMed] [PMC]
    Williams JB, Horton BL, Zheng Y, Duan Y, Powell JD, Gajewski TF. The EGR2 targets LAG-3 and 4-1BB describe and regulate dysfunctional antigen-specific CD8+ T cells in the tumor microenvironment. J Exp Med. 2017;214:381400. [DOI] [PubMed] [PMC]
    Johnson DB, Nixon MJ, Wang Y, Wang DY, Castellanos E, Estrada MV, et al. Tumor-specific MHC-II expression drives a unique pattern of resistance to immunotherapy via LAG-3/FCRL6 engagement. JCI Insight. 2018;3:e120360. [DOI] [PubMed] [PMC]
    Mishra AK, Kadoishi T, Wang X, Driver E, Chen Z, Wang X, et al. Squamous cell carcinomas escape immune surveillance via inducing chronic activation and exhaustion of CD8+ T Cells co-expressing PD-1 and LAG-3 inhibitory receptors. Oncotarget. 2016;7:8134156. [DOI] [PubMed] [PMC]
    Zhang C, Zhang C, Wang H. Immune-checkpoint inhibitor resistance in cancer treatment: Current progress and future directions. Cancer Lett. 2023;562:216182. [DOI] [PubMed]
    Daud AI, Wolchok JD, Robert C, Hwu W, Weber JS, Ribas A, et al. Programmed Death-Ligand 1 Expression and Response to the Anti–Programmed Death 1 Antibody Pembrolizumab in Melanoma. J Clin Oncol. 2016;34:41029. [DOI] [PubMed] [PMC]
    Boyiadzis MM, Kirkwood JM, Marshall JL, Pritchard CC, Azad NS, Gulley JL. Significance and implications of FDA approval of pembrolizumab for biomarker-defined disease. J Immunother Cancer. 2018;6:35. [DOI] [PubMed] [PMC]
    Zhang Z, Lu M, Qin Y, Gao W, Tao L, Su W, et al. Neoantigen: A New Breakthrough in Tumor Immunotherapy. Front Immunol. 2021;12:672356. [DOI] [PubMed] [PMC]
    Peng M, Mo Y, Wang Y, Wu P, Zhang Y, Xiong F, et al. Neoantigen vaccine: an emerging tumor immunotherapy. Mol Cancer. 2019;18:128. [DOI] [PubMed] [PMC]
    Rizvi NA, Hellmann MD, Snyder A, Kvistborg P, Makarov V, Havel JJ, et al. Mutational landscape determines sensitivity to PD-1 blockade in non–small cell lung cancer. Science. 2015;348:1248. [DOI] [PubMed] [PMC]
    Schrörs B, Lübcke S, Lennerz V, Fatho M, Bicker A, Wölfel C, et al. HLA class I loss in metachronous metastases prevents continuous T cell recognition of mutated neoantigens in a human melanoma model. Oncotarget. 2017;8:2831227. [DOI] [PubMed] [PMC]
    Jhunjhunwala S, Hammer C, Delamarre L. Antigen presentation in cancer: insights into tumour immunogenicity and immune evasion. Nat Rev Cancer. 2021;21:298312. [DOI] [PubMed]
    Garris CS, Arlauckas SP, Kohler RH, Trefny MP, Garren S, Piot C, et al. Successful Anti-PD-1 Cancer Immunotherapy Requires T Cell-Dendritic Cell Crosstalk Involving the Cytokines IFN-γ and IL-12. Immunity. 2018;49:114861.e7. Erratum in: Immunity. 2022;55:1749. [DOI] [PubMed] [PMC]
    Ni L, Lu J. Interferon gamma in cancer immunotherapy. Cancer Med. 2018;7:450916. [DOI] [PubMed] [PMC]
    Luo N, Formisano L, Gonzalez-Ericsson PI, Sanchez V, Dean PT, Opalenik SR, et al. Melanoma response to anti-PD-L1 immunotherapy requires JAK1 signaling, but not JAK2. Oncoimmunology. 2018;7:e1438106. [DOI] [PubMed] [PMC]
    Castro F, Cardoso AP, Gonçalves RM, Serre K, Oliveira MJ. Interferon-Gamma at the Crossroads of Tumor Immune Surveillance or Evasion. Front Immunol. 2018;9:847. [DOI] [PubMed] [PMC]
    Sabaawy HE, Ryan BM, Khiabanian H, Pine SR. JAK/STAT of all trades: linking inflammation with cancer development, tumor progression and therapy resistance. Carcinogenesis. 2021;42:14119. [DOI] [PubMed] [PMC]
    Azuma T, Yao S, Zhu G, Flies AS, Flies SJ, Chen L. B7-H1 is a ubiquitous antiapoptotic receptor on cancer cells. Blood. 2008;111:363543. [DOI] [PubMed] [PMC]
    Gato-Cañas M, Zuazo M, Arasanz H, Ibañez-Vea M, Lorenzo L, Fernandez-Hinojal G, et al. PDL1 Signals through Conserved Sequence Motifs to Overcome Interferon-Mediated Cytotoxicity. Cell Rep. 2017;20:181829. [DOI] [PubMed]
    Gao J, Shi LZ, Zhao H, Chen J, Xiong L, He Q, et al. Loss of IFN-γ Pathway Genes in Tumor Cells as a Mechanism of Resistance to Anti-CTLA-4 Therapy. Cell. 2016;167:397404.e9. [DOI] [PubMed] [PMC]
    Shin DS, Zaretsky JM, Escuin-Ordinas H, Garcia-Diaz A, Hu-Lieskovan S, Kalbasi A, et al. Primary Resistance to PD-1 Blockade Mediated by JAK1/2 Mutations. Cancer Discov. 2017;7:188201. [DOI] [PubMed] [PMC]
    Ott PA, Bang Y, Piha-Paul SA, Razak ARA, Bennouna J, Soria J, et al. T-Cell–Inflamed Gene-Expression Profile, Programmed Death Ligand 1 Expression, and Tumor Mutational Burden Predict Efficacy in Patients Treated With Pembrolizumab Across 20 Cancers: KEYNOTE-028. J Clin Oncol. 2019;37:31827. [DOI] [PubMed]
    Karachaliou N, Gonzalez-Cao M, Crespo G, Drozdowskyj A, Aldeguer E, Gimenez-Capitan A, et al. Interferon gamma, an important marker of response to immune checkpoint blockade in non-small cell lung cancer and melanoma patients. Ther Adv Med Oncol. 2018;10:1758834017749748. [DOI] [PubMed] [PMC]
    Ayers M, Lunceford J, Nebozhyn M, Murphy E, Loboda A, Kaufman DR, et al. IFN-γ-related mRNA profile predicts clinical response to PD-1 blockade. J Clin Invest. 2017;127:293040. [DOI] [PubMed] [PMC]
    Karol AB, Fujiwara Y, D’Ovidio T, Baldwin E, Joshi H, Doroshow DB, et al. Peripheral blood cytokines and outcomes with immune checkpoint blockade: a systematic review and meta-analysis. Immunotherapy. 2024;16:82940. [DOI] [PubMed]
    Dufau C, Genais M, Mucher E, Jung B, Garcia V, Montfort A, et al. Ceramide metabolism alterations contribute to Tumor Necrosis Factor-induced melanoma dedifferentiation and predict resistance to immune checkpoint inhibitors in advanced melanoma patients. Front Immunol. 2024;15:1421432. [DOI] [PubMed] [PMC]
    Ardeniz Ö, Unger S, Onay H, Ammann S, Keck C, Cianga C, et al. β2-Microglobulin deficiency causes a complex immunodeficiency of the innate and adaptive immune system. J Allergy Clin Immunol. 2015;136:392401. [DOI] [PubMed]
    Wang H, Liu B, Wei J. Beta2-microglobulin(B2M) in cancer immunotherapies: Biological function, resistance and remedy. Cancer Lett. 2021;517:96104. [DOI] [PubMed]
    Chiou S, Chen C. Decipher β2-microglobulin: gain- or loss-of-function (a mini-review). Med Sci Monit Basic Res. 2013;19:2713. [DOI] [PubMed] [PMC]
    Hicklin DJ, Dellaratta DV, Kishore R, Liang B, Kageshita T, Ferrone S. Beta2-microglobulin gene mutations in human melanoma cells: molecular characterization and implications for immune surveillance. Melanoma Res. 1997;7:S6774. [PubMed]
    Huang L, Malu S, McKenzie JA, Andrews MC, Talukder AH, Tieu T, et al. The RNA-binding Protein MEX3B Mediates Resistance to Cancer Immunotherapy by Downregulating HLA-A Expression. Clin Cancer Res. 2018;24:336676. [DOI] [PubMed] [PMC]
    Petitprez F, Meylan M, Reyniès Ad, Sautès-Fridman C, Fridman WH. The Tumor Microenvironment in the Response to Immune Checkpoint Blockade Therapies. Front Immunol. 2020;11:784. [DOI] [PubMed] [PMC]
    Sharma P, Allison JP. The future of immune checkpoint therapy. Science. 2015;348:5661. [DOI] [PubMed]
    Ribas A, Wolchok JD. Cancer immunotherapy using checkpoint blockade. Science. 2018;359:13505. [DOI] [PubMed] [PMC]
    Tumeh PC, Harview CL, Yearley JH, Shintaku IP, Taylor EJM, Robert L, et al. PD-1 blockade induces responses by inhibiting adaptive immune resistance. Nature. 2014;515:56871. [DOI] [PubMed] [PMC]
    Sade-Feldman M, Yizhak K, Bjorgaard SL, Ray JP, de Boer CG, Jenkins RW, et al. Defining T Cell States Associated with Response to Checkpoint Immunotherapy in Melanoma. Cell. 2018;175:9981013.e20. Erratum in: Cell. 2019;176:404. [DOI] [PubMed] [PMC]
    Tirosh I, Izar B, Prakadan SM, Wadsworth MH 2nd, Treacy D, Trombetta JJ, et al. Dissecting the multicellular ecosystem of metastatic melanoma by single-cell RNA-seq. Science. 2016;352:18996. [DOI] [PubMed] [PMC]
    Lavin Y, Kobayashi S, Leader A, Amir ED, Elefant N, Bigenwald C, et al. Innate Immune Landscape in Early Lung Adenocarcinoma by Paired Single-Cell Analyses. Cell. 2017;169:75065.e17. [DOI] [PubMed] [PMC]
    Zheng C, Zheng L, Yoo J, Guo H, Zhang Y, Guo X, et al. Landscape of Infiltrating T Cells in Liver Cancer Revealed by Single-Cell Sequencing. Cell. 2017;169:134256.e16. [DOI] [PubMed]
    Azizi E, Carr AJ, Plitas G, Cornish AE, Konopacki C, Prabhakaran S, et al. Single-Cell Map of Diverse Immune Phenotypes in the Breast Tumor Microenvironment. Cell. 2018;174:1293308.e36. [DOI] [PubMed] [PMC]
    Guo X, Zhang Y, Zheng L, Zheng C, Song J, Zhang Q, et al. Global characterization of T cells in non-small-cell lung cancer by single-cell sequencing. Nat Med. 2018;24:97885. Erratum in: Nat Med. 2018;24:1628. [DOI] [PubMed]
    Savas P, Virassamy B, Ye C, Salim A, Mintoff CP, Caramia F, et al. Single-cell profiling of breast cancer T cells reveals a tissue-resident memory subset associated with improved prognosis. Nat Med. 2018;24:98693. Erratum in: Nat Med. 2018 ;24:1941. [DOI] [PubMed]
    Zhang L, Yu X, Zheng L, Zhang Y, Li Y, Fang Q, et al. Lineage tracking reveals dynamic relationships of T cells in colorectal cancer. Nature. 2018;564:26872. [DOI] [PubMed]
    Li H, van der Leun AM, Yofe I, Lubling Y, Gelbard-Solodkin D, van Akkooi ACJ, et al. Dysfunctional CD8 T Cells Form a Proliferative, Dynamically Regulated Compartment within Human Melanoma. Cell. 2019;176:77589.e18. Erratum in: Cell. 2020;181:747. [DOI] [PubMed] [PMC]
    Inoue H, Park J, Kiyotani K, Zewde M, Miyashita A, Jinnin M, et al. Intratumoral expression levels of PD-L1, GZMA, and HLA-A along with oligoclonal T cell expansion associate with response to nivolumab in metastatic melanoma. Oncoimmunology. 2016;5:e1204507. [DOI] [PubMed] [PMC]
    Ji R, Chasalow SD, Wang L, Hamid O, Schmidt H, Cogswell J, et al. An immune-active tumor microenvironment favors clinical response to ipilimumab. Cancer Immunol Immunother. 2012;61:101931. [DOI] [PubMed] [PMC]
    Ganesan A, Clarke J, Wood O, Garrido-Martin EM, Chee SJ, Mellows T, et al. Tissue-resident memory features are linked to the magnitude of cytotoxic T cell responses in human lung cancer. Nat Immunol. 2017;18:94050. [DOI] [PubMed] [PMC]
    Gravina AG, Pellegrino R, Esposito A, Cipullo M, Romeo M, Palladino G, et al. The JAK-STAT Pathway as a Therapeutic Strategy in Cancer Patients with Immune Checkpoint Inhibitor-Induced Colitis: A Narrative Review. Cancers (Basel). 2024;16:611. [DOI] [PubMed] [PMC]
    Pellegrino R, Palladino G, Imperio G, Gravina AG. The growing potential of tofacitinib in immune checkpoint inhibitor-induced colitis: identifying remaining puzzle pieces. Explor Immunol. 2024;4:7709. [DOI]
    Luke JJ, Bao R, Sweis RF, Spranger S, Gajewski TF. WNT/β-catenin Pathway Activation Correlates with Immune Exclusion across Human Cancers. Clin Cancer Res. 2019;25:307483. [DOI] [PubMed] [PMC]
    Spranger S, Bao R, Gajewski TF. Melanoma-intrinsic β-catenin signalling prevents anti-tumour immunity. Nature. 2015;523:2315. [DOI] [PubMed]
    Zhang H, Bi Y, Wei Y, Liu J, Kuerban K, Ye L. Blocking Wnt/β-catenin Signal Amplifies Anti-PD-1 Therapeutic Efficacy by Inhibiting Tumor Growth, Migration, and Promoting Immune Infiltration in Glioblastomas. Mol Cancer Ther. 2021;20:130515. [DOI] [PubMed]
    Sun Y, Lu D, Yin Y, Song J, Liu Y, Hao W, et al. PTENα functions as an immune suppressor and promotes immune resistance in PTEN-mutant cancer. Nat Commun. 2021;12:5147. [DOI] [PubMed] [PMC]
    Lin Z, Huang L, Li SL, Gu J, Cui X, Zhou Y. PTEN loss correlates with T cell exclusion across human cancers. BMC Cancer. 2021;21:429. [DOI] [PubMed] [PMC]
    Cachot A, Bilous M, Liu Y, Li X, Saillard M, Cenerenti M, et al. Tumor-specific cytolytic CD4 T cells mediate immunity against human cancer. Sci Adv. 2021;7:eabe3348. [DOI] [PubMed] [PMC]
    Alspach E, Lussier DM, Miceli AP, Kizhvatov I, DuPage M, Luoma AM, et al. MHC-II neoantigens shape tumour immunity and response to immunotherapy. Nature. 2019;574:696701. [DOI] [PubMed] [PMC]
    Ferris ST, Durai V, Wu R, Theisen DJ, Ward JP, Bern MD, et al. cDC1 prime and are licensed by CD4+ T cells to induce anti-tumour immunity. Nature. 2020;584:6249. [DOI] [PubMed] [PMC]
    Kagamu H, Yamasaki S, Kitano S, Yamaguchi O, Mouri A, Shiono A, et al. Single-Cell Analysis Reveals a CD4+ T-cell Cluster That Correlates with PD-1 Blockade Efficacy. Cancer Res. 2022;82:464153. [DOI] [PubMed] [PMC]
    Kryczek I, Zhao E, Liu Y, Wang Y, Vatan L, Szeliga W, et al. Human TH17 cells are long-lived effector memory cells. Sci Transl Med. 2011;3:104ra100. [DOI] [PubMed] [PMC]
    Inomata M, Matsumoto M, Takata N, Hayashi K, Seto Z, Hirai T, et al. Peripheral CD4 memory T cells predict the efficacy of immune checkpoint inhibitor therapy in patients with non-small cell lung cancer. Sci Rep. 2023;13:10807. [DOI] [PubMed] [PMC]
    Bekri S, Rodney-Sandy R, Gruenstein D, Mei A, Bogen B, Castle J, et al. Neoantigen vaccine-induced CD4 T cells confer protective immunity in a mouse model of multiple myeloma through activation of CD8 T cells against non-vaccine, tumor-associated antigens. J Immunother Cancer. 2022;10:e003572. [DOI] [PubMed] [PMC]
    Schildberg FA, Klein SR, Freeman GJ, Sharpe AH. Coinhibitory Pathways in the B7-CD28 Ligand-Receptor Family. Immunity. 2016;44:95572. [DOI] [PubMed] [PMC]
    Kuiper HM, Brouwer M, Linsley PS, van Lier RA. Activated T cells can induce high levels of CTLA-4 expression on B cells. J Immunol. 1995;155:177683. [PubMed]
    Griss J, Bauer W, Wagner C, Simon M, Chen M, Grabmeier-Pfistershammer K, et al. B cells sustain inflammation and predict response to immune checkpoint blockade in human melanoma. Nat Commun. 2019;10:4186. [DOI] [PubMed] [PMC]
    Fässler M, Diem S, Mangana J, Ali OH, Berner F, Bomze D, et al. Antibodies as biomarker candidates for response and survival to checkpoint inhibitors in melanoma patients. J Immunother Cancer. 2019;7:50. [DOI] [PubMed] [PMC]
    Germain C, Gnjatic S, Tamzalit F, Knockaert S, Remark R, Goc J, et al. Presence of B cells in tertiary lymphoid structures is associated with a protective immunity in patients with lung cancer. Am J Respir Crit Care Med. 2014;189:83244. [DOI] [PubMed]
    Murray PJ, Wynn TA. Protective and pathogenic functions of macrophage subsets. Nat Rev Immunol. 2011;11:72337. [DOI] [PubMed] [PMC]
    Russo GL, Moro M, Sommariva M, Cancila V, Boeri M, Centonze G, et al. Antibody-Fc/FcR Interaction on Macrophages as a Mechanism for Hyperprogressive Disease in Non-small Cell Lung Cancer Subsequent to PD-1/PD-L1 Blockade. Clin Cancer Res. 2019;25:98999. [DOI] [PubMed]
    Romano E, Kusio-Kobialka M, Foukas PG, Baumgaertner P, Meyer C, Ballabeni P, et al. Ipilimumab-dependent cell-mediated cytotoxicity of regulatory T cells ex vivo by nonclassical monocytes in melanoma patients. Proc Natl Acad Sci U S A. 2015;112:61405. [DOI] [PubMed] [PMC]
    Gordon SR, Maute RL, Dulken BW, Hutter G, George BM, McCracken MN, et al. PD-1 expression by tumour-associated macrophages inhibits phagocytosis and tumour immunity. Nature. 2017;545:4959. [DOI] [PubMed] [PMC]
    Rakina M, Larionova I, Kzhyshkowska J. Macrophage diversity in human cancers: New insight provided by single-cell resolution and spatial context. Heliyon. 2024;10:e28332. [DOI] [PubMed] [PMC]
    Pathria P, Louis TL, Varner JA. Targeting Tumor-Associated Macrophages in Cancer. Trends Immunol. 2019;40:31027. [DOI] [PubMed]
    Pan Y, Yu Y, Wang X, Zhang T. Tumor-Associated Macrophages in Tumor Immunity. Front Immunol. 2020;11:583084. Erratum in: Front Immunol. 2021;12:775758. [DOI] [PubMed] [PMC]
    Wang D, Yang L, Yue D, Cao L, Li L, Wang D, et al. Macrophage-derived CCL22 promotes an immunosuppressive tumor microenvironment via IL-8 in malignant pleural effusion. Cancer Lett. 2019;452:24453. [DOI] [PubMed]
    Larionova I, Tuguzbaeva G, Ponomaryova A, Stakheyeva M, Cherdyntseva N, Pavlov V, et al. Tumor-Associated Macrophages in Human Breast, Colorectal, Lung, Ovarian and Prostate Cancers. Front Oncol. 2020;10:566511. [DOI] [PubMed] [PMC]
    Shi B, Chu J, Huang T, Wang X, Li Q, Gao Q, et al. The Scavenger Receptor MARCO Expressed by Tumor-Associated Macrophages Are Highly Associated With Poor Pancreatic Cancer Prognosis. Front Oncol. 2021;11:771488. [DOI] [PubMed] [PMC]
    Fleur LL, Boura VF, Alexeyenko A, Berglund A, Pontén V, Mattsson JSM, et al. Expression of scavenger receptor MARCO defines a targetable tumor-associated macrophage subset in non-small cell lung cancer. Int J Cancer. 2018;143:174152. [DOI] [PubMed]
    Georgoudaki A, Prokopec KE, Boura VF, Hellqvist E, Sohn S, Östling J, et al. Reprogramming Tumor-Associated Macrophages by Antibody Targeting Inhibits Cancer Progression and Metastasis. Cell Rep. 2016;15:200011. [DOI] [PubMed]
    Fleur LL, Botling J, He F, Pelicano C, Zhou C, He C, et al. Targeting MARCO and IL37R on Immunosuppressive Macrophages in Lung Cancer Blocks Regulatory T Cells and Supports Cytotoxic Lymphocyte Function. Cancer Res. 2021;81:95667. [DOI] [PubMed]
    Borst J, Ahrends T, Bąbała N, Melief CJM, Kastenmüller W. CD4+ T cell help in cancer immunology and immunotherapy. Nat Rev Immunol. 2018;18:63547. [DOI] [PubMed]
    Guilliams M, Ginhoux F, Jakubzick C, Naik SH, Onai N, Schraml BU, et al. Dendritic cells, monocytes and macrophages: a unified nomenclature based on ontogeny. Nat Rev Immunol. 2014;14:5718. [DOI] [PubMed] [PMC]
    Pesce S, Greppi M, Tabellini G, Rampinelli F, Parolini S, Olive D, et al. Identification of a subset of human natural killer cells expressing high levels of programmed death 1: A phenotypic and functional characterization. J Allergy Clin Immunol. 2017;139:33546.e3. [DOI] [PubMed]
    Pesce S, Greppi M, Grossi F, Zotto GD, Moretta L, Sivori S, et al. PD/1-PD-Ls Checkpoint: Insight on the Potential Role of NK Cells. Front Immunol. 2019;10:1242. [DOI] [PubMed] [PMC]
    Concha-Benavente F, Kansy B, Moskovitz J, Moy J, Chandran U, Ferris RL. PD-L1 Mediates Dysfunction in Activated PD-1+ NK Cells in Head and Neck Cancer Patients. Cancer Immunol Res. 2018;6:154860. [DOI] [PubMed] [PMC]
    Hsu J, Hodgins JJ, Marathe M, Nicolai CJ, Bourgeois-Daigneault M, Trevino TN, et al. Contribution of NK cells to immunotherapy mediated by PD-1/PD-L1 blockade. J Clin Invest. 2018;128:465468. [DOI] [PubMed] [PMC]
    Cui N, Hu M, Khalil RA. Biochemical and Biological Attributes of Matrix Metalloproteinases. Prog Mol Biol Transl Sci. 2017;147:173. [DOI] [PubMed] [PMC]
    Najafi M, Farhood B, Mortezaee K. Extracellular matrix (ECM) stiffness and degradation as cancer drivers. J Cell Biochem. 2019;120:278290. [DOI] [PubMed]
    Théret N, Bouezzedine F, Azar F, Diab-Assaf M, Legagneux V. ADAM and ADAMTS Proteins, New Players in the Regulation of Hepatocellular Carcinoma Microenvironment. Cancers (Basel). 2021;13:1563. [DOI] [PubMed] [PMC]
    Lim JU, Yoon HK. Potential predictive value of change in inflammatory cytokines levels subsequent to initiation of immune checkpoint inhibitor in patients with advanced non-small cell lung cancer. Cytokine. 2021;138:155363. [DOI] [PubMed]
    Chen AY, Wolchok JD, Bass AR. TNF in the era of immune checkpoint inhibitors: friend or foe? Nat Rev Rheumatol. 2021;17:21323. [DOI] [PubMed] [PMC]
    Yi M, Niu M, Zhang J, Li S, Zhu S, Yan Y, et al. Combine and conquer: manganese synergizing anti-TGF-β/PD-L1 bispecific antibody YM101 to overcome immunotherapy resistance in non-inflamed cancers. J Hematol Oncol. 2021;14:146. [DOI] [PubMed] [PMC]
    Leone RD, Emens LA. Targeting adenosine for cancer immunotherapy. J Immunother Cancer. 2018;6:57. [DOI] [PubMed] [PMC]
    Sek K, Mølck C, Stewart GD, Kats L, Darcy PK, Beavis PA. Targeting Adenosine Receptor Signaling in Cancer Immunotherapy. Int J Mol Sci. 2018;19:3837. [DOI] [PubMed] [PMC]
    Schneider E, Winzer R, Rissiek A, Ricklefs I, Meyer-Schwesinger C, Ricklefs FL, et al. CD73-mediated adenosine production by CD8 T cell-derived extracellular vesicles constitutes an intrinsic mechanism of immune suppression. Nat Commun. 2021;12:5911. [DOI] [PubMed] [PMC]
    Munn DH, Mellor AL. IDO in the Tumor Microenvironment: Inflammation, Counter-Regulation, and Tolerance. Trends Immunol. 2016;37:193207. [DOI] [PubMed] [PMC]
    Fujiwara Y, Kato S, Nesline MK, Conroy JM, DePietro P, Pabla S, et al. Indoleamine 2,3-dioxygenase (IDO) inhibitors and cancer immunotherapy. Cancer Treat Rev. 2022;110:102461. [DOI] [PubMed]
    Soldati L, Renzo LD, Jirillo E, Ascierto PA, Marincola FM, Lorenzo AD. The influence of diet on anti-cancer immune responsiveness. J Transl Med. 2018;16:75. [DOI] [PubMed] [PMC]
    Weber DD, Aminzadeh-Gohari S, Tulipan J, Catalano L, Feichtinger RG, Kofler B. Ketogenic diet in the treatment of cancer - Where do we stand? Mol Metab. 2020;33:10221. [DOI] [PubMed] [PMC]
    Ferrere G, Alou MT, Liu P, Goubet A, Fidelle M, Kepp O, et al. Ketogenic diet and ketone bodies enhance the anticancer effects of PD-1 blockade. JCI Insight. 2021;6:e145207. [DOI] [PubMed] [PMC]
    Kian N, Behrouzieh S, Razi S, Rezaei N. Diet Influences Immunotherapy Outcomes in Cancer Patients: A Literature Review. Nutr Cancer. 2023;75:41529. [DOI] [PubMed]
    Prietl B, Treiber G, Pieber TR, Amrein K. Vitamin D and immune function. Nutrients. 2013;5:250221. [DOI] [PubMed] [PMC]
    Golonko A, Pienkowski T, Swislocka R, Orzechowska S, Marszalek K, Szczerbinski L, et al. Dietary factors and their influence on immunotherapy strategies in oncology: a comprehensive review. Cell Death Dis. 2024;15:254. [DOI] [PubMed] [PMC]
    Wolter M, Grant ET, Boudaud M, Steimle A, Pereira GV, Martens EC, et al. Leveraging diet to engineer the gut microbiome. Nat Rev Gastroenterol Hepatol. 2021;18:885902. [DOI] [PubMed]
    Neto AG, Whitaker A, Pei Z. Microbiome and potential targets for chemoprevention of esophageal adenocarcinoma. Semin Oncol. 2016;43:8696. [DOI] [PubMed] [PMC]
    Palm NW, Zoete MRd, Flavell RA. Immune-microbiota interactions in health and disease. Clin Immunol. 2015;159:1227. [DOI] [PubMed] [PMC]
    Shi N, Li N, Duan X, Niu H. Interaction between the gut microbiome and mucosal immune system. Mil Med Res. 2017;4:14. [DOI] [PubMed] [PMC]
    Rooks MG, Garrett WS. Gut microbiota, metabolites and host immunity. Nat Rev Immunol. 2016;16:34152. [DOI] [PubMed] [PMC]
    Al-Asmakh M, Zadjali F. Use of Germ-Free Animal Models in Microbiota-Related Research. J Microbiol Biotechnol. 2015;25:15838. [DOI] [PubMed]
    Geem D, Medina-Contreras O, McBride M, Newberry RD, Koni PA, Denning TL. Specific microbiota-induced intestinal Th17 differentiation requires MHC class II but not GALT and mesenteric lymph nodes. J Immunol. 2014;193:4318. [DOI] [PubMed] [PMC]
    Yoon K, Kim N. The Effect of Microbiota on Colon Carcinogenesis. J Cancer Prev. 2018;23:11725. [DOI] [PubMed] [PMC]
    Cianci R, Pagliari D, Pietroni V, Landolfi R, Pandolfi F. Tissue infiltrating lymphocytes: the role of cytokines in their growth and differentiation. J Biol Regul Homeost Agents. 2010;24:23949. [PubMed]
    Guryanova SV, Ovchinnikova TV. Immunomodulatory and Allergenic Properties of Antimicrobial Peptides. Int J Mol Sci. 2022;23:2499. [DOI] [PubMed] [PMC]
    Jirillo E, Magrone T. Editorial: Antimicrobial Peptides as Mediators of Innate Immunity. Curr Pharm Des. 2018;24:10412. [DOI] [PubMed]
    Miller PL, Carson TL. Mechanisms and microbial influences on CTLA-4 and PD-1-based immunotherapy in the treatment of cancer: a narrative review. Gut Pathog. 2020;12:43. [DOI] [PubMed] [PMC]
    Amemiya K, Meyers JL, Trevino SR, Chanh TC, Norris SL, Waag DM. Interleukin-12 induces a Th1-like response to Burkholderia mallei and limited protection in BALB/c mice. Vaccine. 2006;24:141320. [DOI] [PubMed]
    Peng Z, Cheng S, Kou Y, Wang Z, Jin R, Hu H, et al. The Gut Microbiome Is Associated with Clinical Response to Anti-PD-1/PD-L1 Immunotherapy in Gastrointestinal Cancer. Cancer Immunol Res. 2020;8:125161. [DOI] [PubMed]
    Zhao J, Zhang Z, Xue Y, Wang G, Cheng Y, Pan Y, et al. Anti-tumor macrophages activated by ferumoxytol combined or surface-functionalized with the TLR3 agonist poly (I : C) promote melanoma regression. Theranostics. 2018;8:630721. [DOI] [PubMed] [PMC]
    Davar D, Dzutsev AK, McCulloch JA, Rodrigues RR, Chauvin J, Morrison RM, et al. Fecal microbiota transplant overcomes resistance to anti–PD-1 therapy in melanoma patients. Science. 2021;371:595602. [DOI] [PubMed] [PMC]
    Baruch EN, Youngster I, Ben-Betzalel G, Ortenberg R, Lahat A, Katz L, et al. Fecal microbiota transplant promotes response in immunotherapy-refractory melanoma patients. Science. 2021;371:6029. [DOI] [PubMed]
    Klein A, Friedrich U, Vogelsang H, Jahreis G. Lactobacillus acidophilus 74-2 and Bifidobacterium animalis subsp lactis DGCC 420 modulate unspecific cellular immune response in healthy adults. Eur J Clin Nutr. 2008;62:58493. [DOI] [PubMed]
    Gill HS, Rutherfurd KJ, Prasad J, Gopal PK. Enhancement of natural and acquired immunity by Lactobacillus rhamnosus (HN001), Lactobacillus acidophilus (HN017) and Bifidobacterium lactis (HN019). Br J Nutr. 2000;83:16776. [DOI] [PubMed]
    Tang MLK, Lahtinen SJ, Boyle RJ. Probiotics and prebiotics: clinical effects in allergic disease. Curr Opin Pediatr. 2010;22:62634. [DOI] [PubMed]
    Alsaab HO, Sau S, Alzhrani R, Tatiparti K, Bhise K, Kashaw SK, et al. PD-1 and PD-L1 Checkpoint Signaling Inhibition for Cancer Immunotherapy: Mechanism, Combinations, and Clinical Outcome. Front Pharmacol. 2017;8:561. [DOI] [PubMed] [PMC]
    Ge Z, Peppelenbosch MP, Sprengers D, Kwekkeboom J. TIGIT, the Next Step Towards Successful Combination Immune Checkpoint Therapy in Cancer. Front Immunol. 2021;12:699895. [DOI] [PubMed] [PMC]
    Robert C, Lewis KD, Gutzmer R, Stroyakovskiy D, Gogas H, Protsenko S, et al. Biomarkers of treatment benefit with atezolizumab plus vemurafenib plus cobimetinib in BRAFV600 mutation–positive melanoma. Ann Oncol. 2022;33:54455. [DOI] [PubMed]
    Vinayak S, Tolaney SM, Schwartzberg L, Mita M, McCann G, Tan AR, et al. Open-label Clinical Trial of Niraparib Combined With Pembrolizumab for Treatment of Advanced or Metastatic Triple-Negative Breast Cancer. JAMA Oncol. 2019;5:113240. [DOI] [PubMed] [PMC]
    Obeid M, Tesniere A, Ghiringhelli F, Fimia GM, Apetoh L, Perfettini J, et al. Calreticulin exposure dictates the immunogenicity of cancer cell death. Nat Med. 2007;13:5461. [DOI] [PubMed]
    Zhu H, Shan Y, Ge K, Lu J, Kong W, Jia C. Oxaliplatin induces immunogenic cell death in hepatocellular carcinoma cells and synergizes with immune checkpoint blockade therapy. Cell Oncol (Dordr). 2020;43:120314. [DOI] [PubMed]
    Ye H, Liao W, Pan J, Shi Y, Wang Q. Immune checkpoint blockade for cancer therapy: current progress and perspectives. J Zhejiang Univ Sci B. 2025;26:20326. [DOI] [PubMed] [PMC]
    Formenti SC, Rudqvist N, Golden E, Cooper B, Wennerberg E, Lhuillier C, et al. Radiotherapy induces responses of lung cancer to CTLA-4 blockade. Nat Med. 2018;24:184551. [DOI] [PubMed] [PMC]
    Harding SM, Benci JL, Irianto J, Discher DE, Minn AJ, Greenberg RA. Mitotic progression following DNA damage enables pattern recognition within micronuclei. Nature. 2017;548:46670. [DOI] [PubMed] [PMC]
    Dillon MT, Bergerhoff KF, Pedersen M, Whittock H, Crespo-Rodriguez E, Patin EC, et al. ATR Inhibition Potentiates the Radiation-induced Inflammatory Tumor Microenvironment. Clin Cancer Res. 2019;25:3392403. [DOI] [PubMed] [PMC]
    Sevenich L. Turning “Cold” Into “Hot” Tumors—Opportunities and Challenges for Radio-Immunotherapy Against Primary and Metastatic Brain Cancers. Front Oncol. 2019;9:163. [DOI] [PubMed] [PMC]
    Viallard C, Larrivée B. Tumor angiogenesis and vascular normalization: alternative therapeutic targets. Angiogenesis. 2017;20:40926. [DOI] [PubMed]
    Pan C, Liu H, Robins E, Song W, Liu D, Li Z, et al. Next-generation immuno-oncology agents: current momentum shifts in cancer immunotherapy. J Hematol Oncol. 2020;13:29. [DOI] [PubMed] [PMC]
    Yi M, Jiao D, Qin S, Chu Q, Wu K, Li A. Synergistic effect of immune checkpoint blockade and anti-angiogenesis in cancer treatment. Mol Cancer. 2019;18:60. [DOI] [PubMed] [PMC]
    Anderson TS, Wooster AL, Piersall SL, Okpalanwaka IF, Lowe DB. Disrupting cancer angiogenesis and immune checkpoint networks for improved tumor immunity. Semin Cancer Biol. 2022;86:98196. [DOI] [PubMed] [PMC]
    Rahma OE, Hodi FS. The Intersection between Tumor Angiogenesis and Immune Suppression. Clin Cancer Res. 2019;25:544957. [DOI] [PubMed]
    Wu J, Zhao X, Sun Q, Jiang Y, Zhang W, Luo J, et al. Synergic effect of PD-1 blockade and endostar on the PI3K/AKT/mTOR-mediated autophagy and angiogenesis in Lewis lung carcinoma mouse model. Biomed Pharmacother. 2020;125:109746. [DOI] [PubMed]
    Zhao S, Ren S, Jiang T, Zhu B, Li X, Zhao C, et al. Low-Dose Apatinib Optimizes Tumor Microenvironment and Potentiates Antitumor Effect of PD-1/PD-L1 Blockade in Lung Cancer. Cancer Immunol Res. 2019;7:63043. [DOI] [PubMed]
    Li Q, Wang Y, Jia W, Deng H, Li G, Deng W, et al. Low-Dose Anti-Angiogenic Therapy Sensitizes Breast Cancer to PD-1 Blockade. Clin Cancer Res. 2020;26:171224. [DOI] [PubMed]
    Creelan BC, Wang C, Teer JK, Toloza EM, Yao J, Kim S, et al. Tumor-infiltrating lymphocyte treatment for anti-PD-1-resistant metastatic lung cancer: a phase 1 trial. Nat Med. 2021;27:14108. [DOI] [PubMed] [PMC]
    Bai X, Zhou Y, Yokota Y, Matsumoto Y, Zhai B, Maarouf N, et al. Adaptive antitumor immune response stimulated by bio-nanoparticle based vaccine and checkpoint blockade. J Exp Clin Cancer Res. 2022;41:132. [DOI] [PubMed] [PMC]
    Chen Q, Sun T, Jiang C. Recent Advancements in Nanomedicine for ‘Cold’ Tumor Immunotherapy. Nanomicro Lett. 2021;13:92. [DOI] [PubMed] [PMC]