- Review
- Open access
- Published:
Current trends in sensitizing immune checkpoint inhibitors for cancer treatment
Molecular Cancer volume 23, Article number: 279 (2024)
Abstract
Immune checkpoint inhibitors (ICIs) have dramatically transformed the treatment landscape for various malignancies, achieving notable clinical outcomes across a wide range of indications. Despite these advances, resistance to immune checkpoint blockade (ICB) remains a critical clinical challenge, characterized by variable response rates and non-durable benefits. However, growing research into the complex intrinsic and extrinsic characteristics of tumors has advanced our understanding of the mechanisms behind ICI resistance, potentially improving treatment outcomes. Additionally, robust predictive biomarkers are crucial for optimizing patient selection and maximizing the efficacy of ICBs. Recent studies have emphasized that multiple rational combination strategies can overcome immune checkpoint resistance and enhance susceptibility to ICIs. These findings not only deepen our understanding of tumor biology but also reveal the unique mechanisms of action of sensitizing agents, extending clinical benefits in cancer immunotherapy. In this review, we will explore the underlying biology of ICIs, discuss the significance of the tumor immune microenvironment (TIME) and clinical predictive biomarkers, analyze the current mechanisms of resistance, and outline alternative combination strategies to enhance the effectiveness of ICIs, including personalized strategies for sensitizing tumors to ICIs.
Introduction
Immune checkpoint inhibitors (ICIs) block regulatory pathways that suppress T cell activity and immune responses, leading to robust activation of the immune system [1,2,3,4]. Mechanistically, cancer cells can exploit these immune checkpoints to evade host immune surveillance, offering a potential strategy for cancer immunotherapy [5,6,7,8,9,10]. The successful clinical development of ICIs, such as CTLA-4 and PD-1/PD-L1 inhibitors, has revolutionized cancer treatment by delivering unprecedented anti-tumor activity. This progress has facilitated the clinical use of ICIs as monotherapies or in combination with other therapies [11,12,13,14,15,16,17,18]. Since the approval of ipilimumab, a CTLA-4-targeting monoclonal antibody, in 2011 for advanced metastatic melanoma, ICIs have become a standard treatment for several solid tumors, including those with microsatellite instability-high (MSI-H) tumors [19,20,21,22,23,24,25]. Numerous immune checkpoints are currently being investigated for their translational potential, from preclinical studies to clinical trials, based on promising preclinical results [26,27,28,29,30,31,32]. For example, relatlimab, a recently FDA-approved drug targeting the lymphocyte-activation gene 3 (LAG-3) protein, is used in combination with nivolumab for treating unresectable or metastatic melanoma [33,34,35]. Ongoing research is focused on exploring novel immune checkpoints and developing rational therapeutic combinations with synergistic anti-tumor mechanisms to expand the indications and clinical benefits of ICIs.
Despite the significant clinical benefits observed in some patients, with notable tumor regression in response to ICIs, a substantial proportion of patients either do not respond to these treatments or exhibit low response rates [36,37,38,39]. Some patients experience innate resistance or develop acquired resistance over time, which diminishes the effectiveness of ICIs [40,41,42]. Currently, numerous clinical trials are investigating various combination therapies with ICIs as the core component to enhance treatment sensitivity [15, 43,44,45]. However, few strategies have yielded encouraging clinical results so far [46,47,48]. Understanding the mechanisms underlying both resistance and response to ICIs is crucial for predicting immune response outcomes and potentially converting non-responders into responders. Furthermore, it is essential to incorporate robust biological principles and prospective biomarkers when evaluating different drug combinations for patient screening [43, 49,50,51,52,53,54,55]. This review explores the mechanisms and factors involved in immune checkpoint response and resistance, assesses clinically integrated immunotherapy response biomarkers, and highlights recent advances in immunotherapy combination strategies aimed at expanding the clinical applications of ICIs.
Immune checkpoint inhibitors (ICIs) in cancer immunotherapy
Over the past decade, advances in tumor immunology have significantly improved our understanding of the immune system’s role in cancer treatment. This has led to the development of immunoregulatory monoclonal antibodies targeting T-cell immune checkpoints such as CTLA-4 and PD-1, which have revitalized anti-tumor immunotherapy and cancer precision medicine. Additionally, antibodies against other immune checkpoint molecules like LAG3, TIM3, TIGIT, ICOS, GITR, and 4-1BB are actively being explored in clinical trials. This section focuses on the mechanisms of CTLA-4 and PD-1 in T-cell-mediated anti-tumor immunity and reviews key clinical trials related to these treatments.
Programmed cell death protein 1 (PD-1)
Programmed death 1 (PD-1, CD279) was identified in 1992 as a key regulator of peripheral tolerance. It is expressed on activated/ exhausted T cells, as well as on B cells, natural killer (NK) cells, macrophages, dendritic cells (DCs), and immature Langerhans’ cells [56, 57]. PD-1 functions through its ligands, PD-L1 and PD-L2, by inhibiting T-cell receptor (TCR) signaling. This inhibition occurs through the phosphorylation of immunoreceptor tyrosine-based switch motifs (ITSM) and the recruitment of Src homology region 2 domain-containing phosphatase-1 (SHP-1) and - 2 (SHP-2), which dephosphorylate ZAP70 and CD3ζ, impairing TCR activation [30, 58,59,60,61,62]. Additionally, PD-1 signaling downregulates the PI3K-Akt-mTOR and RAS-MEK-ERK pathways. Specifically, PTEN phosphorylation, regulated by CK2, suppresses PI3K signaling, while inhibition of Ras and PLC-γ1 affects the RAS-MEK-ERK pathway [63,64,65,66,67,68,69]. PD-1/PD-L1 interactions also inhibit B-cell responses by affecting the phosphorylation of signaling molecules such as PI3K and Syk [70, 71]. Mechanistically, CTLA-4 acts early in T cell activation to block CD28-B7 interactions, downregulating antigen-presenting cells (APCs)-induced T cell responses and primarily affecting CD4+ T cell amplification and trafficking. In contrast, PD-1 is expressed later in T cell activation and regulates the effective stage of T cell response by restraining TCR signaling, with anti-PD-1/PD-L1 therapy primarily enhancing the antitumor immunity of exhausted CD8+ T cells. Thus, the distinct mechanisms of CTLA-4 and PD-1 highlight their nonoverlapping roles in T cell functional inhibition, supporting the potential benefits of combinatorial targeted strategies [72,73,74,75,76,77,78].
Over the past few years, several pivotal trials have reshaped the treatment landscape. In a phase III trial comparing pembrolizumab to ipilimumab in advanced melanoma, pembrolizumab demonstrated superior progression-free and 12-month overall survival without high-grade toxicity (KEYNOTE-006, NCT01866319) [79]. This finding was reinforced by subsequent analyses, leading to pembrolizumab’s first approval for the treatment of advanced melanoma, particularly after initial treatment with ipilimumab and BRAF inhibitors [80,81,82]. Nivolumab, another PD-1 inhibitor, first entered clinical trials in 2006 and showed promising results in a 2012 phase I trial for refractory solid tumors. In phase III trials, nivolumab exhibited durable objective responses and lower toxicity compared to other chemotherapy regimens in advanced melanoma patients who had progressed after previous treatments. A separate phase III trial also showed nivolumab’s superior overall survival (OS) compared to dacarbazine in treatment-naive advanced melanoma patients, with survival benefits linked to PD-L1 expression [83,84,85]. Combining CTLA-4 and PD-1 inhibitors has shown promise in metastatic melanoma, with a phase III trial revealing that combination therapy significantly improved progression-free survival (PFS) compared to either nivolumab or ipilimumab monotherapy alone, even in PD-L1 negative tumors patients [84]. This led to the FDA’s 2014 approval of nivolumab for melanoma treatment. Advances in the field of immune checkpoint blocking therapy for melanoma have facilitated the exploration of ICI in other tumor types. For example, in non-small cell lung cancer (NSCLC), pembrolizumab outperformed traditional chemotherapy in improving PFS and OS, leading to its FDA approval as a first-line treatment for NSCLC with high PD-L1 expression [86]. Currently, several anti-PD-1/PD-L1 therapies have proven beneficial in NSCLC, small cell lung cancer (SCLC), gastric cancer (GC), hepatocellular carcinoma (HCC), colorectal cancer (CRC), and bladder cancer, and many more are currently under investigation [87,88,89,90,91].
Cytotoxic T lymphocyte-associated protein 4 (CTLA-4)
Cytotoxic T lymphocyte antigen-4 (CTLA-4) is a type I transmembrane protein primarily expressed on T cells and upregulated upon T cell activation. It plays a crucial role in regulating T cell responses early in activation, as independently demonstrated in the mid-1990s [92,93,94,95]. CTLA-4 suppresses T cell activation through both intrinsic and extrinsic mechanisms. CTLA-4 binds with high affinity to B7 molecules, competing with CD28 for binding to B7 family members on APCs, thereby blocking the CD28-B7 signaling pathway and preventing CD28-mediated T cell activation [96]. Additionally, CTLA-4 exerts negative regulation by inhibiting IL-2 production and IL-2 receptor expression, suppressing T cell proliferation and activation by blocking the transition from G1 to S phase, and disrupting TCR signaling through interactions with PP2A and SHP-2 [97,98,99,100,101]. CTLA-4 is constitutively expressed on regulatory T cells (Tregs) that maintain self-tolerance, CTLA-4-knockout Tregs fail to regulate autoimmunity, leading to severe immune hyperactivation and premature death in mice [102,103,104]. These mechanisms collectively enable CTLA-4 to play a critical role in immune tolerance.
Initially, two fully humanized CTLA-4-directed monoclonal antibodies, ipilimumab (MDX-010) and tremelimumab, were developed and tested in clinical trials for advanced melanoma starting in 2000 [11, 105]. Ipilimumab was first evaluated in single-dose trials for patients with metastatic melanoma, ovarian cancer (OC), and prostate cancer [106,107,108,109,110,111,112]. Subsequent multi-dose studies showed that administering ipilimumab at 3 mg/kg every 3 weeks, combined with peptide vaccines, significantly improved OS and tumor regression compared to peptide vaccines alone [113]. This established the importance of CTLA-4 in cancer immunotherapy and led to a surge in immuno-oncology research. In 2010, a pivotal phase III trial demonstrated that ipilimumab provided a 3.5-month median OS benefit over the gp100 peptide vaccine alone, leading to its FDA approval in 2011 for late-stage melanoma [106, 114]. However, tremelimumab, another anti-CTLA-4 mAb, did not show a significant survival advantage in several late-phase trials. For instance, a phase III trial comparing tremelimumab to standard chemotherapy for metastatic melanoma failed to show significant improvement [115]. These setbacks led to the exploration of combination therapies [116, 117]. Phase III HIMALAYA trail combining tremelimumab with durvalumab showed promising results, including a potent immune response and enhanced anti-tumor activity in HCC (NCT03298451) [118]. Based on these findings, the combination was approved by the FDA in October 2022 for unresectable hepatocellular carcinoma (uHCC) and in November 2022 for metastatic non-small cell lung cancer (mNSCLC) [105, 116, 119]. Despite the progress, many combination trials still fail to meet primary OS endpoints [120].
Other immune checkpoints
Apart from CTLA-4 and PD-1, several positive and negative immune checkpoint molecules have been identified and investigated over the past decade as potential immunotherapeutic targets [121,122,123].
LAG-3 is highly expressed on activated T cells and various other immune cells, where it binds to MHC class II and other ligands, delivering inhibitory signals that suppress T cell activity and enhance Treg-mediated immunosuppression [124]. Several studies have demonstrated that LAG-3 and PD-1 synergize on CD8 + T cells to drive T cell exhaustion and impede autocrine IFN-γ-dependent anti-tumor immunity [125]. This synergy provides insight into how combinatorial targeting of LAG-3 and PD-1 can enhance therapeutic efficacy. Subsequent clinical studies have confirmed these synergistic effects. For instance, the RELATIVITY-047 trial, a global, double-blind, randomized Phase 2/3 study, showed improved PFS in patients with metastatic or unresectable melanoma when treated with a combination of relatlimab and nivolumab, compared to PD-1 inhibition alone [33]. Other clinical studies, including those on head and neck squamous cell carcinoma (HNSCC), GC and lung cancer, are ongoing (NCT04811027, CTR20221603) [126].
TIM-3 is a co-inhibitory receptor expressed on a range of immune cells, including IFN-γ-secreting CD4 + and CD8 + T cells, NK cells, myeloid cells, mast cells, Tregs, and B cells. It interacts with ligands such as galectin-9, phosphatidylserine, high-mobility group protein B1, and carcinoembryonic antigen cell adhesion molecule-1 to suppress anti-tumor immunity via inducing terminal dysfunction and death of CD8+ T cells [127,128,129,130]. A range of anti-TIM-3 agents have been developed based on preclinical data and effects observed in advanced cancer patients, with sabatolimab and cobolimab being the most advanced. Several clinical trials are ongoing for anti-TIM-3 therapies across hematologic malignancies and solid tumors [131,132,133,134,135]. Notably, sabatolimab in combination with hypomethylating agents has shown durable responses, earning Fast-Track designation from the FDA and orphan drug status from the European Commission for MDS [136]. In a Phase I study, cobolimab combined with dostarlimab (PD-1 inhibitor) achieved a 42.9% objective response rate (ORR) in NSCLC patients without severe adverse events (AEs). Cobolimab is now being tested in combination with chemotherapy (docetaxel) and/or anti-PD-1 in various cancers [137].
TIGIT is a newly identified co-inhibitory receptor that suppresses T cell proliferation and cytokine production by interacting with its ligands CD155 and CD112. Clinical trials investigating anti-TIGIT monoclonal antibodies, both alone and in combination with anti-PD-1 and anti-PD-L1 therapies, have shown promising early results [138,139,140,141]. Notably, the CITYSCAPE-02 trial demonstrated that tiragolumab (TIGIT inhibitor) plus atezolizumab improved overall response rates in PD-L1-positive, recurrent or metastatic NSCLC, earning FDA breakthrough therapy designation [139]. In addition, the MORPHEUS-liver study showed that tiragolumab combined with atzolizumab and bevacizumab improved ORR and FPS in first-line liver cancer patients [142]. Despite these advancements, the phase III SKYSCRAPER-02 trial found no clinical benefit for TIGIT targeting in SCLC [143]. Overall, while anti-TIGIT therapies have demonstrated notable progress in specific indications, the results across different studies have been mixed [144].
Co-stimulatory checkpoint molecules are being explored for cancer immunotherapy in both preclinical and clinical studies. These molecules, found not only on T cells but also on other immune cells, enhance anti-tumor immune responses. Immune co-stimulator (ICOS), expressed on T cells, is induced by T cell activation and facilitates T cell differentiation and expansion. Several clinical trials are currently investigating the effects of targeting ICOS alone or in combination with ICIs (NCT05695898, NCT02520791, NCT04319224) [145, 146].
Checkpoint molecules of the tumor necrosis factor receptor (TNFR) superfamily, including 4-1BB, OX40, and glucocorticoid-induced TNFR-related gene (GITR), are being investigated as therapeutic targets [147,148,149,150]. 4-1BB, also known as CD137 or TNFRSF9, is a transmembrane protein upregulated upon T cell activation by mitogens or antigens. Engagement of 4-1BB with its exclusive ligand 4-1BBL forms a hexameric complex, which enhances the transcription of anti-apoptotic genes in T cells, thereby promoting long-lasting memory responses of cytotoxic CD8+ T cells. The overexpression of 4-1BBL and administration of agonistic monoclonal antibodies targeting 4-1BB have been shown to improve CD8+ T cell-mediated anti-tumor responses and induce tumor rejection in preclinical models. This underscores the potential of incorporating 4-1BB signaling domains into CAR-T cell constructs to enhance activation and therapeutic efficacy [151]. OX40, expressed transiently on T cells after activation, enhances T cell expansion and memory formation upon interaction with its ligand OX40L. It also modulates Tregs by inhibiting their function while promoting their production, thereby playing a complex role in anti-tumor immunity [152]. Several agonist antibodies targeting OX40 and GITR are currently being explored in preclinical and clinical trials [153,154,155,156,157,158].
The complex tumor immune microenvironment is a highly promising biomarkers for tumor immunotherapy
Immune checkpoint inhibitors response and TIME
The discovery of immune checkpoints has revolutionized cancer treatment, significantly improving clinical outcomes and overall survival. Following the FDA approval of ipilimumab in 2011, interest in immunotherapy surged. Over the past decade, several monoclonal antibodies targeting PD-1 and PD-L1 have also received FDA approval for various cancers, including melanoma, NSCLS, renal cell carcinoma (RCC), HNSCC, GC, HCC, and more [19, 159, 160]. Despite these advancements, resistance to immune checkpoint therapy (ICT) remains common, categorized into primary resistance (no initial response) and acquired resistance (initial response followed by relapse). Some cancers, like pancreatic cancer and glioblastoma, show inherent resistance, while others, such as melanoma and bladder cancer, may develop resistance after initial success [161,162,163,164]. Understanding the mechanisms of resistance is crucial for predicting patient responses and improving treatment strategies. Though robust biomarkers are lacking, analyzing the tumor microenvironment (TME) and immune cell composition pre- and post-therapy may reveal predictive markers for effective ICT. The cancer-immunity cycle highlights the role of T cells in anti-tumor immunity, from activation in lymph nodes to tumor cell elimination [165]. However, tumors often exploit immune checkpoints like PD-1 to evade detection, leading to T-cell exhaustion. Immunotherapy aims to reverse this exhaustion and enhance immune responses.
T cells play a central role in anti-tumor immunity, guided by the classical cancer-immunity cycle. This cycle outlines how APCs, such as DCs, process tumor antigens and present them as major histocompatibility complex (MHC)-peptide complexes to T cell receptors (TCRs). This interaction triggers T cell priming and activation, regulated by co-stimulatory and inhibitory checkpoints in lymph nodes, leading to T cell proliferation, enhanced effector functions, and memory formation [166]. Once activated, T cells migrate to the TME to target and destroy tumor cells. However, tumors often exploit inhibitory checkpoint molecules like PD-1 to evade T cell-mediated destruction. This results in T-cell exhaustion, a state of diminished immune response due to the immunosuppressive TME [167, 168]. Immunotherapy aims to counteract this exhaustion by reactivating T cells and improving their ability to combat tumors. Effective therapies often focus on reversing T cell exhaustion and enhancing anti-tumor immune responses to limit tumor progression and promote tumor elimination.
The complex interactions between the host’s immune system and cancer cells within the TME significantly influence immune cell functions [169]. Understanding these interactions is crucial for deciphering how tumors evade immune detection. The concept of “immune contexture” refers to the diverse immune variables, including cell types, densities, functional states, and distributions within the TME, which can indicate survival outcomes and predict treatment responses [170, 171]. The TME is classified into three categories (hot, altered and cold) based on immune activity and tumor escape mechanisms. Hot Tumors have high lymphocyte infiltration and PD-1 expression, often showing good responses to ICIs. They are characterized by high levels of tumor-infiltrating lymphocytes (TILs), PD-L1 expression on immune cells, and high genomic instability. In contrast, cold tumors show low levels of lymphocyte infiltration and minimal PD-L1 expression. They typically have low antigen presentation capabilities, such as reduced major histocompatibility complex class I (MHC I) expression, and low mutational burden, making them less responsive to ICT. This classification provides a framework to assess tumor immune profiles and predict responses to immunotherapy, potentially guiding more effective treatment strategies [1, 172, 173].
Clinically integrated biomarkers of ICI response
Efforts to predict responses to cancer immunotherapy and guide personalized treatment have intensified. Identifying and standardizing early predictive biomarkers related to immunotherapy sensitivity and resistance are crucial for optimizing ICB therapies. Proposed and tested biomarkers include immune cell infiltration, Immunoscore, tumor mutational burden (TMB), neoantigen load, PD-L1 expression, mismatch repair deficiency (dMMR), microsatellite instability (MSI), and inflammatory gene profiling within the TME [49, 174]. These biomarkers aim to identify the most effective therapeutic interventions for individual patients. Here, immunotherapy biomarker applied for the oncology clinic will be delineated.
PD-L1 expression
PD-L1 expression in total nucleated cells (both malignant and immune cells in the TME), as detected by immunohistochemistry (IHC), is the most commonly used predictive biomarker for selecting patients for ICIs therapy. High PD-L1 expression often correlates with better responses to PD-1/PD-L1 axis blockade, making it a valuable tool for identifying patients who might achieve longer survival [175, 176]. In clinical practice, PD-L1 expression is evaluated using four FDA-approved IHC assays (22C3, 28-8, SP263, and SP142), each validated through rigorous staining processes. Pre-treatment PD-L1 expression is assessed using scoring systems such as the combined positive score (CPS), tumor proportion score (TPS), and immune cell score (IC). Scoring methods and cut-off values for predicting immunotherapy response vary across clinical trials and tumor types (e.g., gastric or lung cancer). Currently, PD-L1 status has associated with regulatory approvals for several cancers, including NSCLC, GC/GEJC, bladder cancer, and cervical cancer [177, 178]. However, PD-L1 status is only predictive in about 30% of cases across these cancer types, with its clinical utility varying by cancer type and ICI agent [178]. Some patients with PD-L1-negative tumors also show responses, and phase III trials have not always shown a clear correlation between PD-L1 expression and survival outcomes. This variability underscores the challenge of using PD-L1 as a standalone biomarker [179]. Furthermore, differences in tissue handling techniques, detecting antibody, immune cell staining heterogeneity, and assay sensitivity complicate the standardization of PD-L1 testing. Therefore, improving standardization and integrating PD-L1 with other biomarkers could enhance predictive accuracy.
Tumor mutation burden (TMB) and MSI
Tumor mutation burden (TMB), defined as the number of non-synonymous mutations per megabase of coding sequence, is being investigated as a predictive biomarker for ICI therapy. High TMB can generate tumor neoantigens (mutation-associated neoantigens, or MANAs) that may be recognized by the immune system, potentially triggering an effective antitumor response. Advances in next-generation sequencing and molecular characterization have enhanced our understanding of cancer biology and facilitated precision medicine. Whole exome sequencing (WES) is considered the gold standard for measuring TMB in tumor biopsies [180, 181]. Clinical studies have shown that higher TMB is associated with better outcomes in ICI therapy, leading to the FDA’s approval of TMB as a companion diagnostic for pembrolizumab in treating pediatric and adult patients with unresectable or metastatic solid tumors with high TMB (≥ 10 mutations/megabase) [182]. Notably, not all mutations are detectable by the immune system, and a high TMB does not always ensure a positive response to ICT, nor does a low TMB rule out an effective immune response. This makes TMB an imperfect standalone biomarker across different tumor types [183]. Additionally, technical and biological limitations, such as the lack of standardization and a universal threshold for defining high TMB, complicate its use. The commonly used threshold of 10 mutations per megabase is constrained by variability across cancer types, making it difficult to apply universally. Future research should focus on validating TMB thresholds and exploring the immunogenicity of various mutation patterns [184]. The analysis of WES data from both germline and tumor DNA is complex, and limited availability of WES data in diagnostic biopsies further hinders its widespread use. In heterogeneous tumors with many subclonal mutations, detecting mutations becomes less effective as tumor purity decreases. These challenges may be addressed through advancements in bioinformatics, targeted next-generation sequencing, and machine learning-based TMB detection [185, 186].
In addition, genome damage due to deficiencies in the DNA mismatch repair system is closely linked to immunogenicity because it results in a high mutational load. DNA damage repair (DDR) defects can lead to increased TMB and more neoantigen formation [187, 188]. For example, tumors with high microsatellite instability (MSI-H) or dMMR show greater sensitivity to ICIs than those with proficient mismatch repair (pMMR). Initial studies found that CRC patients with dMMR had significantly better responses to pembrolizumab compared to those with pMMR status [189]. Subsequent clinical trials confirmed the utility of MSI as a predictive biomarker for immunotherapy, leading to the FDA’s approval of pembrolizumab for MSI-H/dMMR solid tumors and nivolumab for MSI-H CRC.
ICI sensitization strategies and clinical research progress
Based on the increasing understanding of the mechanisms underlying ICB resistance, research is focused on combining ICBs with various therapeutic strategies to enhance treatment sensitivity and efficacy (Fig. 1). Additionally, the ongoing major clinical trials on sensitizing ICIs for cancer therapy are summarized in Table 1.
Immune checkpoint combinations
Several ICIs have been approved by the FDA, showing significant clinical benefits across various cancers. However, the response rate of ICIs varies by cancer type, ranging from 10 to 45%, prompting the investigation of combination therapies to improve efficacy [190]. CTLA-4 inhibitors primarily enhance DCs activity, which is suppressed by Tregs, and activate antigen-specific CD4+ T cells in the lymph nodes. This boosts T cell priming, expands T cell diversity, and reduces Tregs in the TME [191]. Conversely, PD-1/PD-L1 inhibitors aim to reverse T cell exhaustion and activate NK cells in tumors and lymph nodes, restoring cytotoxic T lymphocytes (CTLs) function. Combining CTLA-4 and PD-1/PD-L1 inhibitors leverages these distinct mechanisms, showing promising clinical results [192]. Therefore, this dual approach has demonstrated higher ORR and improved OS compared to monotherapy in melanoma, CRC, NSCLC, HCC, and RCC.
For instance, the CheckMate 067 trial revealed that nivolumab combined with ipilimumab significantly improved median OS and PFS in advanced melanoma [193]. Similarly, CheckMate 204 showed durable responses and long-term benefits for patients with melanoma brain metastases when treated with nivolumab plus ipilimumab [194]. The CheckMate 227 trial found that this combination improved 5-year survivorship compared to chemotherapy, regardless of PD-L1 expression [195]. Additionally, the CheckMate 142 study demonstrated that nivolumab plus low-dose ipilimumab was well-tolerated and effective as a first-line treatment for MSI-H/dMMR mCRC [196, 197]. Moreover, in the neoadjuvant setting, this combination achieved an impressive 100% pathological response rate in early-stage dMMR colon cancers [197].
While combination therapies offer enhanced efficacy, not all patients benefit equally. Ongoing research is exploring third-generation ICIs with non-overlapping mechanisms, such as targeting LAG-3, TIGIT, TIM-3, VISTA, and BTLA. Recent trials, such as those with relatlimab plus nivolumab and tiragolumab plus atezolizumab, have shown encouraging results, though some have not yet demonstrated significant improvements in PFS or OS [33, 139, 198]. Multiple trials are currently investigating these and other combination strategies to further improve outcomes (NCT05645692, NCT05785767, NCT04811027, NCT05483400, NCT04305054).
Combination with chemotherapy
Cytotoxic chemotherapy remains a cornerstone in cancer treatment, effective for most malignancies. It rapidly reduces tumor burden and induces immunogenic cell death (ICD), which can enhance immune responses by releasing cancer antigens for potential presentation. This effect can complement ICIs, potentially improving treatment outcomes [199, 200]. Chemotherapy’s benefits include stimulating tumor-specific immunity and synergistically enhancing ICI efficacy. For instance, combining chemotherapy with ICIs has shown success in treating NSCLC, SCLC, GC, biliary tract cancer, and triple-negative breast cancer (TNBC) [201,202,203,204,205,206]. Chemotherapy promotes the release of damage-associated molecular patterns (DAMPs) from tumor cells, which activate DCs and enhance antigen presentation. This process improves T cell activation and reduces the presence of immunosuppressive cells like Tregs, myeloid-derived suppressor cells (MDSCs), and M2 macrophages in the TME [207, 208]. It also encourages the differentiation of tumor-associated macrophages (TAMs) into the M1 phenotype, boosting both innate and adaptive immunity.
Several chemotherapy and ICI combinations have received regulatory approval [209]. The KEYNOTE-189 trial demonstrated that adding pembrolizumab to standard chemotherapy (pemetrexed-platinum) significantly improved response rates and OS in untreated metastatic non-squamous NSCLC, with a 48.3% ORR compared to 19.9% for chemotherapy alone. Long-term results from this study showed a 3-year OS of 71.9% among patients receiving pembrolizumab. Similarly, the CheckMate 816 trial found that neoadjuvant Opdivo plus platinum-based chemotherapy significantly extended median event-free survival and increased the rate of pathological complete response (pCR) in resectable NSCLC [210, 211]. In the KEYNOTE-355 study, pembrolizumab combined with chemotherapy notably improved PFS in metastatic TNBC with high PD-L1 expression (CPS ≥ 10), leading to accelerated FDA approval [212]. Numerous ongoing clinical trials are investigating additional combination regimens and their effects on various cancers (NCT0276357, NCT02578680, NCT02366143, NCT02872116, NCT02746796, NCT03138512, NCT03838159, NCT02488759) [205, 213,214,215,216,217]. Despite these advances, only a subset of patients benefits from combination therapies. Identifying biomarkers to better target these treatments remain crucial for optimizing patient outcomes [51, 218,219,220,221].
Combination with radiotherapy
Radiotherapy (RT) is a cornerstone of cancer treatment, with emerging evidence showing it can have both tumor-promoting and tumor-suppressing immune effects. RT induces significant DNA damage, ICD, and tumor shrinkage, which can enhance immune responses by generating neoantigens and increasing the release of inflammatory cytokines. This promotes the infiltration of effector CD8+ T cells and DCs into the TME, potentially leading to an abscopal effect on non-irradiated lesions [222]. However, RT also has immunosuppressive effects, such as inducing anti-immunogenic cytokines and increasing the presence of immunosuppressive cells like Tregs, MDSCs, and M2 macrophages [223]. Recent studies have found that RT can upregulate PD-L1 expression on DCs and myeloid cells, which may facilitate metastasis. Blocking the PD-L1/CXCL10 axis or MDSC infiltration during local irradiation can mitigate these effects [224].
Combining RT with ICIs shows potential synergy. For instance, the Phase III PACIFIC trial demonstrated that durvalumab, an anti-PD-L1 antibody, significantly improved PFS and OS in stage III NSCLC patients after chemoradiotherapy [87, 225,226,227]. Similarly, adding durvalumab to neoadjuvant therapy with stereotactic body radiotherapy (SBRT) increased the MPR in early NSCLC [228]. In metastatic NSCLC, combining SBRT with pembrolizumab improved response rates, though not all studies have met predefined efficacy criteria [229, 230]. Despite mixed results in other cancers, such as HNSCC and Merkel cell carcinoma, ongoing research aims to optimize RT and ICI combinations by adjusting treatment timing, dose, and site. Low-dose radiation strategies are being explored as potential enhancers of ICI response [231,232,233,234,235].
Preclinical studies suggest low-dose RT can enhance immune checkpoint inhibitor effectiveness by promoting macrophage differentiation into the M1 phenotype and increasing T cell recruitment [231, 232, 236]. For example, combining low-dose RT with dual PD-L1 and VEGFA blockers has shown promise in treating HCC by activating exhausted CD8+ T cells [237]. Additionally, a recent study indicated that low-dose RT combined with immunotherapy can significantly improve outcomes in SCLC patients, with high ORR and prolonged PFS [235]. Overall, while combining RT with immunotherapy holds promise, further research is needed to identify optimal protocols and patient subgroups to maximize therapeutic benefits.
Combination with targeted therapy
Cancer is a genetic disease driven by multiple mutations. Targeted therapies, focusing on molecules involved in cancer processes such as carcinogenesis, angiogenesis, and metastasis, have emerged as crucial components of precision medicine. These therapies target specific molecules that support tumor progression, offering a clinical basis for mechanism-based treatment of malignancies. Key therapeutic targets include BRAF, EGFR, HER2, MEK, and PARP. Many targeted inhibitors have been approved by the FDA, either as monotherapies or in combination with other drugs, showing promising clinical results [238, 239]. Preclinical studies indicate that targeting tumor signaling pathways can enhance T cell infiltration, restore dendritic cell function, neutralize MDSCs, and improve antigen presentation, providing a rationale for combining ICBs with targeted therapies [240].
The BRAF oncogene, present in many malignant melanomas and other cancers, is a major focus of targeted drug discovery. BRAF and MEK inhibitors are first-line treatments for BRAFV600 mutation-positive melanoma, and combinations with immunotherapy are being actively studied (NCT02967692; NCT02752074; NCT02625337; NCT03178851; NCT02908672) [241,242,243,244,245]. The Phase III IMspire150 study found that adding atezolizumab to BRAF and MEK inhibitors (vemurafenib and cobimetinib) significantly improved PFS (15.1 months vs. 10.6 months) in BRAFV600 mutation-positive melanoma [246, 247]. This combination also showed activity in patients with CNS metastases [248].
Sequencing immunotherapy and targeted therapy is being explored to optimize combination efficacy and identify biomarkers for predicting long-term benefits [249,250,251,252,253,254]. Additionally, anti-PD-1/PD-L1 therapy has shown comparable efficacy to other targeted therapies such as PARP inhibitors, MERTK inhibitors, PI3K inhibitors, and CDK4/6 inhibitors, which are being tested for improved responses to ICBs [255,256,257,258,259].
Combination with anti-angiogenic therapy
Aberrant tumor angiogenesis, a hallmark of cancer, significantly contributes to tumor immunosuppression and immune evasion. Tumor blood vessel abnormalities can cause intra-tumor hypoxia (ITH) and a low pH TIME, which are linked to decreased T cell infiltration [260,261,262]. Additionally, vascular endothelial growth factor (VEGF) and angiopoietin-2 (ANGPT2) influence the TIME by inhibiting DCs maturation, upregulating PD-L1 on endothelial cells, and promoting the polarization of TAMs into immunosuppressive M2-like phenotypes [263].
Anti-angiogenic therapies, including anti-VEGF agents (bevacizumab), anti-VEGFR agents (ramucirumab), and tyrosine kinase inhibitors (sorafenib, lenvatinib, apatinib, sunitinib, axitinib), work by disrupting neovascularization and limiting tumor nutrient supply. This leads to improved vascular perfusion, increased immune cell infiltration, and reduced presence of MDSCs, Tregs, and M2 macrophages in the TIME, thereby enhancing anti-tumor effects [264]. Emerging preclinical and clinical evidence indicates that combining ICBs with anti-angiogenic therapies during the “vascular normalization” phase can improve clinical outcomes [265]. In various preclinical models, VEGFR inhibitors combined with ICBs have demonstrated synergistic effects by regulating neo-angiogenesis, reversing ITH, and re-sensitizing tumors to ICB therapy.
Clinical trials have confirmed the benefits of this combination approach. The Phase III KEYNOTE-426 study showed that the combination of pembrolizumab and axitinib resulted in significantly longer OS and PFS compared to sunitinib in untreated advanced clear-cell RCC, establishing a new standard of care [266]. Similarly, a Phase II trial is evaluating nivolumab plus axitinib in anti-PD-1 refractory unresectable advanced melanoma [267]. In addition, the Phase III IMbrave150 study led to the FDA approval of atezolizumab plus bevacizumab in 2020 as a first-line treatment for unresectable or metastatic HCC [268]. Numerous ongoing Phase III trials are investigating this combination therapy across various cancers, with initial results showing promising anti-tumor activity (NCT03764293, NCT03468218, NCT04047017, NCT03950154, NCT04547088, NCT03141177) [269,270,271,272,273,274].
Combination with adoptive T cell therapy
Combining ICIs with adoptive T cell therapy is an area of significant interest. Adoptive T cell therapy involves large infusions of tumor-specific T lymphocytes isolated from the TIME. These T cells are specific to tumor antigens and can bypass defects in endogenous antigen delivery, making them a promising strategy for stimulating anti-tumor immune responses.
Chimeric antigen receptor (CAR) T cells are engineered T cells with receptors specific to tumor antigens. The CAR fusion protein, comprising an antibody binding domain and a T cell receptor signaling domain, is introduced into autologous T cells using lentivirus or retrovirus. CAR-T cells can recognize tumor-specific cell surface antigens without MHC restrictions, allowing for precise targeted cytotoxicity and expanding therapeutic potential applications. Preclinical and clinical studies have demonstrated that CAR-T cell therapies can provide durable and meaningful responses, particularly in hematological malignancies [275]. However, the complexity of the TIME in solid tumors and upregulation of inhibitory immune checkpoints can impair CAR-T cell function. Combining CAR-T cell therapy with ICBs may enhance anti-tumor efficacy by overcoming these challenges [276].
Studies have shown that CAR-T cells with endogenous PD-1 checkpoint blockade exhibit improved anti-tumor activity [277,278,279,280]. For instance, Rafiq et al. engineered CAR-T cells to secrete a PD-1 blocking single chain variable fragment (scFv), which enhanced the anti-tumor activity of CAR-T cells in a xenogeneic mouse model [281]. Currently, the mechanism of blocking the PD1/PD-L1 axis to reverse the function of CAR-T cells is also under intensive investigation. Recent studies have also shown that PD-L1 upregulation on M2 macrophages significantly inhibits CAR-T cell activity. Combining PD L1 blockade with CAR-T cell therapy has been shown to promote M1-like macrophages and reduce M2 macrophages, thereby improving CAR-T cell function [282]. Moreover, downregulation of PD-1 expression on CAR-T cells can enhance their anti-tumor efficacy by preserving early cellular memory and reducing T cell depletion [283,284,285,286]. CAR-T cells combined with PD-1/PD-L1 axis blockade have shown promising results in preclinical studies across various cancers, including breast cancer, TNBC, advanced thyroid cancer, CRC, and glioblastoma [287,288,289,290]. Clinical trials are actively exploring these combinations [291, 292]. For example, a Phase Ib study of a novel anti-CD19 CAR expressing PD-1/CD28 chimeric converter receptor demonstrated high remission rates in patients with PD-L1-positive B-cell lymphoma [293]. Similarly, a Phase I study of mesothelin-targeted CAR- T cells combined with PD-1 blockade showed potent anti-tumor activity and long-term CAR-T cell survival in malignant pleural mesothelioma patients [294]. Similarly, a Phase I study of mesothelin-targeted CAR-T cells combined with PD-1 blockade showed potent anti-tumor activity and long-term CAR-T cell survival in malignant pleural mesothelioma patients [295].
In summary, combining CAR-T cell therapy with PD-1/PD-L1 blockade has shown significant potential in preclinical and clinical studies, with ongoing research aimed at optimizing these strategies for improved outcomes in cancer treatment [296,297,298,299,300,301].
Combination with oncolytic viruses
Oncolytic viruses (Ovs) offer a promising therapeutic approach that combines direct tumor cell destruction with the induction of systemic anti-tumor immunity. These viruses selectively replicate within tumor cells, leading to cell lysis and the release of tumor-specific antigens, which in turn stimulates both innate and adaptive immune responses. Additionally, Ovs can be engineered to enhance their immunostimulatory effects, thereby amplifying pro-inflammatory responses in the TIME and improving immune-mediated tumor elimination at both local and distant sites [302]. Given the challenges associated with tumor resistance to immunotherapy, combining Ovs with ICBs is being explored as a potentially synergistic strategy to boost immune responses. This combination approach has been extensively studied in preclinical and clinical trials.
Talimogene Laherparepvec (T-VEC), an attenuated herpes simplex virus type 1 (HSV-1), is the first FDA-approved oncolytic viral therapy. It is used to treat skin and lymph node lesions in patients with unresectable melanoma [303, 304]. T-VEC is genetically engineered to express GM-CSF, which enhances anti-tumor responses by promoting DCs maturation and stimulating tumor antigen-reactive T cells. In a Phase II study, T-VEC combined with ipilimumab showed a significantly higher persistent ORR compared to ipilimumab alone in unresectable stage IIIB/IV melanoma patients, without additional safety concerns. This was the first randomized controlled trial to assess the addition of an oncolytic virus to a checkpoint inhibitor and achieved its primary endpoint [305, 306]. However, a subsequent global Phase III trial found that combining T-VEC with pembrolizumab did not significantly improve PFS or OS compared to pembrolizumab alone, despite promising Phase Ib results [307].
RP1 is another HSV-1-based oncolytic virus that encodes a fusogenic protein (GALV-GP-R-) and expresses human GM-CSF. In a Phase 1/2 trial, RP1 combined with nivolumab demonstrated promising anti-tumor activity in patients with cutaneous cancers, including those with anti-PD-1 refractory stage IIIb/IV melanoma. Tumor biopsies from treated patients showed evidence of immune activation, such as increased CD8+ T cell infiltration and heightened PD-L1 expression [308]. Initial data from an expanded cohort suggest that RP1 plus nivolumab continues to provide durable and clinically significant anti-tumor activity in patients with progressive melanoma, with an overall ORR of 36.3% [309].
Currently, multiple clinical trials are investigating various Ovs in combination with ICBs (NCT05733611, NCT06311578, NCT05076760, NCT05346484, NCT06067061, NCT05222932, NCT05564897, NCT05271318, NCT04725331), showing promising potential for this combined therapeutic strategy [310,311,312,313,314,315,316,317,318,319].
Combination with vaccine
Cancer immunotherapy has transformed oncology, offering effective treatments across various cancers. Understanding the cancer-immune cycle is crucial for grasping the mechanisms and sensitivities of anti-cancer immune responses. DCs in tumor-draining lymph nodes capture antigens and present them to naive T cells, initiating and sustaining anti-tumor immunity. Therapeutic cancer vaccines aim to stimulate the immune system against tumors by combining antigens with adjuvants to activate DCs and induce a lasting anti-tumor memory [320]. Various vaccine platforms are in clinical trials, including DC-based, DNA-based, RNA-based, peptide-based, and virus-based vaccines [321]. Successful vaccines often target specific antigens, which may be either shared antigens overexpressed in tumors or neoantigens arising from somatic mutations. Current research is focusing on neoantigen-targeted vaccines to improve specificity and minimize off-target effects. However, challenges like low TMB and neoantigen immunogenicity complicate vaccine development [322]. Sipuleucel-T, a DC-based vaccine approved for hormone-refractory prostate cancer, has shown limited survival benefits. To enhance efficacy, combining vaccines with ICBs is being explored to overcome immunosuppressive mechanisms and improve vaccine performance [323, 324].
In recent trials, combining a personalized therapeutic cancer vaccine (PTCV) with pembrolizumab showed promising results in advanced HCC, with an ORR of 30.6% and a complete response (CR) of 8.3%. Neoantigen-specific T cell responses and increased T cell infiltration at the tumor site were observed [18]. Additionally, an ongoing trial involving a vaccine encoding 20 shared neoantigens, utilizing self-amplified mRNA (samRNA) and chimpanzee adenovirus (ChAd68), in combination with ipilimumab and nivolumab, has demonstrated acceptable tolerance and potential efficacy in advanced solid tumors [325]. Future advancements in vaccine platforms and TIME profiling will further enhance the efficacy of these combined therapies [245, 326,327,328,329,330,331].
Combined with FMT
A growing body of preclinical and clinical evidence indicates that the gut microbiome can significantly impact the efficacy of cancer immunotherapy. Advances in high-throughput sequencing technologies have enabled extensive research into how gut microbiota influences the host immune system. Studies have demonstrated that the microbial genome affects hormone secretion, metabolism, immune function, and physiological homeostasis through various mechanisms [332, 333]. For instance, Matson et al. identified a notable correlation between the composition of gut commensal microorganisms and the clinical response of patients with metastatic melanoma by analyzing baseline fecal samples before immunotherapy. They observed that bacterial species such as Bifidobacterium longum, Enterococcus faecalis, and Collinsella globosa were more prevalent in patients who responded to ICIs. Moreover, fecal microbiota transplantation (FMT) from these responders into germ-free mice enhanced T-cell responses and improved the efficacy of anti-PD-L1 therapy [334]. Similarly, Routy et al. found that the abundance of Akkermansia muciniphila was associated with clinical responses to ICIs in cancer patients. Their macro-genomic analysis of fecal samples at diagnosis revealed that higher levels of this bacterium correlated with better treatment outcomes. Furthermore, increased recruitment of CCR9+ CXCR3+ CD4+ T lymphocytes within tumors and the restoration of PD-1 blockade efficacy were observed in an interleukin-12-dependent manner. A meta-analysis of melanoma patients treated with PD-1 inhibitors further supported the complex relationship between gut microbiota and immunotherapy responses [335,336,337]. These findings underscore the potential of gut microbiome biomarker analysis to enhance immune regulation and address tumor immune checkpoint resistance, particularly following antibiotic pretreatment [338, 339].
Molecular mechanisms underlying tumor sensitization to immunotherapy
To expand the clinical benefits of ICIs, it is crucial to understand the mechanisms of drug resistance and response to ICB. While significant progress has been made in understanding ICI resistance, further exploration of sensitization mechanisms is needed. This section reviews the genomic and molecular factors, both intrinsic and extrinsic to tumors, that are associated with resistance and response to tumor immunotherapy (Fig. 2).
Tumor intrinsic mechanisms of sensitization to ICI
Antigen-presenting molecules machinery: (MHC, HLA)
Cytotoxic CD8+ T lymphocytes initiate effective anti-tumor immunity by recognizing tumor antigens presented by MHC I. However, defects in the neoantigen presentation machinery, mediated by MHC I downregulation due to chromosomal deletions or loss of heterozygosity (LOH), are considered as primary or acquired resistance mechanisms that enable evasion of T cell recognition [340,341,342]. MHC I molecules, consisting of membrane-anchored heavy chains and β2-microglobulin (β2m), are essential for antigen presentation. Mutations in β2m or LOH can reduce MHC I expression on tumor cells, facilitating immune evasion. Additionally, epigenetic alterations, such as chromatin modifications, abnormal DNA methylation, and histone changes, can silence MHC I expression, further promoting immune escape [343,344,345]. Understanding the mechanisms by which tumor cells evade immune elimination is crucial for enhancing the effectiveness of cancer immunotherapy. MHC-I expression is regulated by IFNGR signaling, which involves STAT1-mediated induction of the IRF1 transcription factor that binds to MHC-I gene promoters. In optineurin-deficient CRC cells, impaired IFNγ and MHC-I signaling leads to palmitoylation-dependent lysosomal sorting and degradation of IFNGR1, resulting in decreased MHC-I levels and intrinsic resistance to immunotherapy. This presents a potential pharmacological strategy to sensitize checkpoint therapy in CRC. Additionally, post-transcriptional regulatory mechanisms also impact MHC-I expression. NBR1-mediated macroautophagy can selectively degrade MHC-I molecules, facilitating immune evasion in pancreatic ductal adenocarcinoma (PDAC). This degradation can be reversed by autophagy inhibition with chloroquine, either genetically or pharmacologically [346, 347]. By degrading MHC molecules, tumor cells reduce their visibility to immune cells, particularly cytotoxic T cells, which rely on MHC for recognition and targeting of cancer cells. NK cells can target cells lacking MHC I, but tumors often evade this by reducing MICA/B levels on their surface, presenting a significant challenge for current immunotherapies.
Immunogenic neoantigens: mismatch repair (pMMR/dMMR)
Genomic and chromosomal instability is a hallmark of cancer, altering the genomic landscape and influencing immune responses. Tumors with non-synonymous mutations or genomic rearrangements often produce highly immunogenic neoantigens. Defects in DNA repair pathways, such as the mismatch repair (MMR) system, increase the formation of clonal neoantigens, making tumors more recognizable and sensitive to ICIs [197, 348]. The MMR system, primarily involving the proteins MLH1, MSH2, MSH6, and PMS2, corrects replication errors not fixed by DNA polymerase proofreading. Germline mutations in MMR genes, such as in Lynch syndrome, lead to MSI due to the accumulation of point mutations and defects in repeat sequences. Tumors with MSI-H or dMMR display frequent frameshift mutations and higher TMB, making them more responsive to ICIs [349,350,351]. The increased mutation load stimulates the generation of immunogenic neoantigens, which triggers T cell infiltration and immune activation. However, immune editing can lead to the loss of these neoantigens by selectively removing highly immunogenic tumor cell subpopulations, representing a mechanism of acquired resistance to immune surveillance.
Epigenetic reprogramming
Epigenetics involves heritable changes in gene expression that do not alter the DNA sequence. Key mechanisms include DNA methylation, chromatin remodeling, histone modification, and non-coding RNA regulation [352]. These processes ensure precise and lasting gene regulation. Tumorigenesis often features significant genetic and epigenetic alterations. For example, about 20% of cancers exhibit loss-of-function mutations in genes encoding the SWI/SNF chromatin remodeling complex [353]. DNA methylation, primarily regulated by DNA methyltransferase 1 (DNMT1), typically leads to gene silencing and is a stable feature of cells, often disrupted in cancer. Tumor cells exploit epigenetic mechanisms to evade immune surveillance and reduce the efficacy of ICB, often by suppressing endogenous interferon responses. By modifying the epigenetic landscape, tumor cells can either activate or repress human endogenous retroviruses (HERVs), influencing immune responses. Activation of HERVs can trigger an interferon-related innate immune response, enhancing tumor cell detection through viral mimicry. Chromatin-modifying agents, such as DNA methyltransferase inhibitors (DNMTis), can reactivate HERVs, inducing a type I interferon response and promoting apoptosis in OC models. Additionally, DNMTis and histone deacetylase inhibitors (HDACis) can generate HERV-derived immunogenic neoantigens, increasing tumor cell visibility to cytotoxic T cells and boosting immune-mediated tumor destruction [354,355,356,357].
Epigenetic changes can also drive tumor-specific neoantigen formation and reactivation of developmental genes, producing differentiation antigens [358, 359]. Loss of MHC-I expression in tumors may be due to the epigenetic silencing of antigen-presenting genes, such as TAP1, TAP2, and β2m. However, this can be reversed with DNMTis or HDACis. Additionally, global DNA methylation can upregulate PD-L1 and inhibitory cytokines. The cGAS-STING signaling pathway, which detects abnormal DNA and triggers type I interferon responses, is often inhibited by epigenetic silencing in tumors, reducing immunotherapy efficacy. In preclinical models, DNMT1 and EZH2-mediated modifications inhibit T-helper 1 (Th1) chemokines CXCL9 and CXCL10, blocking effector T cell infiltration and correlating with poor clinical outcomes in cancers like OC [360,361,362]. Understanding these epigenetic mechanisms offers potential therapeutic strategies to overcome immunosuppression and enhance immunotherapy effectiveness.
Oncogenic signaling pathways activation
Tumor intrinsic oncogenic signaling pathways drive malignant progression and contribute to the maintenance of an immunosuppressive TME. Mutations in pathways such as IFNγ/JAK/STAT, WNT/β-catenin, PTEN/PI3K, and RAS have been shown to both induce immunogenic responses and mediate immune exclusion, thereby fostering immune resistance. Investigating how these oncogenic pathways influence resistance to ICIs may offer valuable insights for enhancing the effectiveness of immune checkpoint therapies.
IFNγ/JAK/STAT signaling pathway
Type II interferon (IFN-γ), primarily secreted by CD8+ CTLs, is crucial for anti-tumor immunity. When IFN-γ binds to its receptors IFNGR1 and IFNGR2, it activates Janus kinases (JAK1 and JAK2) and the STAT signaling pathway. This activation enhances gene transcription and antigen presentation, bolstering anti-tumor effects and increasing cytotoxic T cell infiltration into tumors [363, 364]. However, IFN-γ/JAK/STAT signaling can also contribute to ICI resistance. This occurs when interferon regulatory factor 1 (IRF1) drives the transcriptional upregulation of PD-L1, leading to PD-L1-independent resistance. Mutations in the JAK-STAT pathway, such as loss-of-function mutations in JAK1/JAK2, can impair IFN-γ’s efficacy and serve as potential predictors of ICB resistance [365].
WNT/β-catenin
The Wnt signaling pathway regulates cell homeostasis and stem cell function. Mutation-driven activation of the Wnt-β-catenin pathway is common in cancers, promoting self-renewal and contributing to resistance to ICB therapies [366, 367]. Abnormal Wnt/β-catenin activation induces upregulation of the transcription factor ATF3, leading to immune ignorance in the TME. This pathway inhibits the transcription of chemokine CCL4, reducing the recruitment of CD103+ DCs and impeding T cell infiltration. Consequently, this results in CD4+ T cell tolerance and CD8+ T cell immunosuppression due to ineffective cross-priming with DCs [368,369,370]. Analysis from The Cancer Genome Atlas (TCGA) reveals that Wnt/β-catenin activation correlates with non-T cell inflammatory gene expression across various tumors [371,372,373]. Given its role in immunosuppression, targeting the Wnt/β-catenin pathway is being explored as a strategy to overcome ICB resistance.
PI3K-AKT-mTOR
The PI3K-AKT-mTOR signaling pathway is crucial for cell growth, proliferation, and survival. Mutations activating this pathway are prevalent in various cancers and often lead to increased PD-L1 expression on tumor cells due to PTEN or PIK3CA gene alterations, potentially enabling tumor immune evasion and affecting sensitivity to ICB therapy. PTEN, a frequently inactivated tumor suppressor gene, negatively regulates the PI3K-AKT-mTOR pathway, contributing to anti-tumor activity. PTEN loss is associated with decreased T cell infiltration and reduced expression of oncolytic cytokine genes. This loss is more common in non-T-cell inflammatory tumors, which exhibit less response to PD-1 inhibitors compared to PTEN wild-type tumors [374,375,376]. Conversely, in preclinical models of PTEN-mutated prostate cancer and melanoma, reactivating PTEN through mRNA nanoparticle technology has shown promise in enhancing immune activation and anti-tumor efficacy when combined with anti-PD1 therapy [377]. These findings suggest that targeting the PI3K-AKT-mTOR pathway and PTEN could improve ICB therapy outcomes, supporting the exploration of combination strategies involving PI3K inhibitors.
RAS
RAS genes (KRAS, NRAS, HRAS) are commonly mutated oncogenes that encode small GTP-binding proteins crucial for linking upstream receptors to downstream signaling pathways involved in cell survival, proliferation, migration, and metabolism. Mutations in RAS lock the protein in an active GTP-bound state, leading to continuous activation of downstream pathways such as RAF-MEK-ERK and PI3K-AKT [378, 379]. Clinical trials have shown that KRAS mutations, particularly in NSCLC, are associated with increased PD-L1 expression, which promotes tumor immune escape by stabilizing PD-L1 mRNA. Additionally, RAS pathway alterations can reduce IFNγ-induced MHC expression and affect T cell infiltration in cancers like colorectal and breast cancer. Preclinical studies suggest that combining KRAS G12C inhibitors with anti-PD-1 therapy enhances CD8+ T cell infiltration and improves immunotherapy sensitivity [380,381,382,383,384].
Immunological extrinsic mechanisms for sensitizing to ICIs in the tumor microenvironment
Tumor extracellular matrix (ECM) components and cytokines
The TME consists of immune and non-immune components, such as stromal cells, blood vessels, cytokines, and extracellular matrix, which influence tumor characteristics like angiogenesis, invasion, and metastasis. These components interact with tumor and immune cells, significantly affecting tumor progression and resistance to ICB therapies [385]. The tumor-promoting microenvironment is characterized by abundant immunosuppressive myeloid cells, dense desmoplastic stroma that obstructs cytotoxic T-cell infiltration and systemic chemotherapy, and an increase in M2 macrophages. Remodeling the TME with CD40 agonists can polarize M2 macrophages to M1, activate APCs, enhance tumor-specific T-cell responses, degrade fibrotic stroma, and improve chemotherapy sensitivity. In clinical practice, the combination of the CD40 agonist mitazalimab with the mFOLFIRINOX regimen effectively activates immune cells and promotes anti-tumor responses, presenting a promising therapeutic option for metastatic PDAC with poor prognosis [386].
Epithelial-Mesenchymal Transition (EMT) enables epithelial cells to acquire mesenchymal traits, decreasing their susceptibility to immune-mediated lysis. EMT also contributes to immune desensitization through mechanisms such as altered PD-L1/PD-L2 expression, reduced sensitivity to cell death receptor pathways, and immune synaptic defects [387,388,389]. High angiogenesis can lead to abnormal blood vessels and increased interstitial pressure, hindering immune cell infiltration and ICB efficacy.
Cancer-associated fibroblasts (CAFs), influenced by signals like TGF-β, Wnt/β-catenin, PI3K-AKT-mTOR, JAK/STAT, and EGFR, play a complex role in the TME. They promote tumor growth, immune evasion, and resistance to therapy [390,391,392]. TGF-β signaling, which can be activated through canonical Smad pathways or non-Smad pathways (e.g., MAPK, Rho GTPases, PI3K-Akt-mTOR), affects ECM gene expression and immune cell composition, potentially creating an immunologically “hot” TME [393,394,395]. However, CAFs expressing fibroblast-activating protein (FAP) can suppress anti-tumor T cell functions, especially in gastrointestinal tumors. Additionally, TGF-β impairs therapeutic sensitivity of cetuximab by up-regulating CD59 and inhibits T cell proliferation which further reducing ICB therapy susceptibility [396, 397].
Immune cell
Myeloid cells are key components of the TIME and are linked to poor prognosis and reduced efficacy of immunotherapy across various cancers. MDSCs, a crucial myeloid subgroup, are known for their immunosuppressive effects, which contribute to resistance to ICIs. MDSCs release mediators like arginase 1, peroxynitrite, inducible nitric oxide synthase (iNOS), and reactive oxygen species (ROS), creating an immunosuppressive TME [398]. This downregulates T cell and NK cell activity, affects Tregs differentiation, and induces an immunosuppressive macrophage phenotype, reducing immunotherapy effectiveness. Tumor-infiltrating MDSCs also express high levels of PD-L1 and other inhibitory checkpoints. Elevated PI3K-γ expression in myeloid cells can further exacerbate inflammation and tumor progression. In preclinical models, targeting PI3K-γ has been shown to restore ICI sensitivity by reprogramming macrophages and enhancing T cell activity [399].
Tumor-associated macrophages (TAMs), another major myeloid subset, play a significant role in tumor immune regulation. TAMs are categorized into M1 and M2 phenotypes. M1 macrophages promote anti-tumor responses with pro-inflammatory cytokines, whereas M2 macrophages suppress CD8+ T cell activation through anti-inflammatory cytokines, recruit Tregs, and facilitate tumor immune escape [400]. M2 TAMs also upregulate inhibitory checkpoint molecules, amplifying their immunosuppressive effects. Inhibiting M2-TAM activity and shifting macrophages toward the M1 phenotype can counteract ICIs resistance [401].
Regulatory T cells (Tregs), characterized by CD25, CD4, and FoxP3, are highly immunosuppressive and crucial for maintaining immune tolerance [402]. Tregs are prevalent in the TIME and bloodstream, where they diminish CD4+ and CD8+ T cell responses through cytokine secretion (e.g., IL-2, IL-10, IL-35, TGF-β) and checkpoint molecule expression (e.g., CTLA-4, PD-1). Tregs create an immunosuppressive environment by secreting inhibitory cytokines and expressing CTLA-4 and PD-1, which further downregulates co-stimulatory molecules on APCs and increases free PD-L1 [403]. Preclinical studies suggest that depleting tumor-infiltrating Tregs can enhance anti-tumor immunity and ICIs response [404]. However, incomplete depletion may lead to Treg expansion and the upregulation of alternative checkpoints like TIM-3 and LAG-3, contributing to ICB resistance.
Discussion
Immunotherapy has emerged as a crucial pillar in cancer treatment, with significant advances in ICIs marking its progress. Despite notable successes in inducing tumor regression and prolonging disease control across various cancer types, many patients either do not respond to these therapies or develop resistance over time. Understanding the mechanisms behind such resistance and incorporating clinical translational research are essential for improving outcomes and optimizing patient selection through biomarker-based strategies. Recent advancements in genomics, transcriptomics, and other molecular analyses have facilitated the identification of predictive markers for ICI response and resistance. High-throughput sequencing technologies are particularly valuable for dissecting the complex mechanisms underlying treatment resistance. While these technologies offer insights into genomic and functional alterations that contribute to resistance, identifying robust prognostic biomarkers remains a challenge. Broadly exploratory biomarkers have shown promise in guiding clinical trials, yet precise, validated markers are needed for effective patient stratification. Intra- and inter-tumoral heterogeneity complicates the analysis of immune responses, and preclinical models using identical transplantable cell lines often fail to replicate clinical realities. To address this, researchers are developing advanced preclinical tumor models using patient-derived samples that preserve TIME signatures. These models help elucidate both immune and non-immune pathways influencing anti-tumor immunity and provide a better understanding of factors affecting immune response beyond the tumor itself.
Currently, strategies to enhance the efficacy of tumor immunotherapy mainly focus on combining different immune checkpoint inhibitors, targeted therapies (especially anti-angiogenic agents), radiotherapy (including different doses and various radiotherapy modalities), and chemotherapy. New treatment approaches, such as the combination with CAR-T therapy, also play a crucial role in cancer treatment. In recent years, several studies have found high expression of immune-suppressive molecules like LAG3 and IDO in PD-L1+ populations, and combined inhibition of LAG-3 and PD-L1 can significantly enhance the activity of effector T cells. Recent research indicates that combination immunotherapy with PD-L1/LAG3 dual-target agents has shown promising clinical response rates in cancers such as esophageal and liver cancer. In addition to LAG3, TIGIT and TIM3 have also been confirmed by multiple studies to co-express with PD-1/PD-L1, and the co-expression of PD-L1/TIGIT and PD-L1/TIM3 is associated with differences in OS. Therefore, shifting from single-target to dual-target approaches might be a strategy to overcome primary resistance to immunotherapy. Currently, the most researched strategy involves using preemptive anti-angiogenic therapy to reverse immune resistance. Regorafenib and apatinib combined with PD-1 monoclonal antibodies have shown good efficacy. In addition, the approach of combining immunotherapy with the blockade of driver genes such as KRAS, MYC, EGFR, and HER2 to reverse resistance is also receiving considerable attention.
While various immune sensitization methods exist, future clinical practice must tailor treatment strategies based on patients’ multi-omics profiles (e.g., genomics, transcriptomics, immunomics, metabolomics) and clinical features (e.g., tumor pathological type, stage, patient age). The goal is to personalize therapies by understanding each patient’s unique clinical and molecular characteristics, optimizing efficacy while minimizing side effects. Despite the potential of personalized immune sensitization, several challenges remain in its clinical application: (1) tumor microenvironment complexity: tumor evolution and intratumor heterogeneity (ITH) drive disease progression and resistance to ICIs. Advances in single-cell analysis and spatial transcriptomics are improving our understanding of TME heterogeneity, enabling novel strategies like cellular immunotherapy. Technologies such as scRNA-seq, scATAC-seq, and whole-genome sequencing provide deeper insights into TME dynamics and resistance pathways; (2) analyzing multi-omics data is resource-intensive and demands advanced technical support and specialized expertise. However, the integration of machine learning and AI models is crucial for advancing the field. Recent advances in combining neural network-based machine learning with multi-omics data may improve predictions of responses to immuno-chemotherapy, helping to address this challenge [405,406,407]; (3) managing side effects in combination therapies: the combination of ICIs with other treatments can lead to increased side effects, making it crucial to balance efficacy with safety. In conclusion, as multi-omics research and AI technologies continue to advance, personalized strategies for sensitizing tumors to ICIs can lead to the development of highly effective treatment plans that maximize immunotherapy outcomes. Personalized immune sensitization is poised to become a cornerstone of cancer treatment.
As the mechanisms of resistance to immune checkpoint therapy are further unraveled, there will be critical advancements in clinical translational research. This progress will drive the development of more effective, personalized immune-oncology strategies, including rational combinations of immune checkpoint blockers with other therapeutic modalities. By integrating these insights, we can enhance the sensitivity of immunotherapy and improve clinical outcomes for individual patients.
Data availability
No datasets were generated or analysed during the current study.
Abbreviations
- ICIs:
-
Immune Checkpoint Inhibitors
- ICB:
-
Immune Checkpoint Blockade
- TIME:
-
Tumor Immune Microenvironment
- MSI-H:
-
Microsatellite Instability-High
- LAG-3:
-
Lymphocyte-Activation Gene 3
- APCs:
-
Antigen-Presenting Cells
- NSCLC:
-
Non-Small Cell Lung Cancer
- SCLC:
-
Small Cell Lung Cancer
- samRNA:
-
self-amplified mRNA
- VEGF:
-
Vascular Endothelial Growth Factor
- ANGPT2:
-
Angiopoietin-2
- Tregs:
-
regulatory T cells
- uHCC:
-
unresectable Hepatocellular Carcinoma
- HCC:
-
Hepatocellular Carcinoma
- mNSCLC:
-
metastatic Non-Small Cell Lung Cancer
- OS:
-
Overall Survival
- PFS:
-
Progression-Free Survival
- ICOS:
-
Immune Co-Stimulator
- TNFR:
-
Tumor Necrosis Factor Receptor
- GITR:
-
Glucocorticoid-Induced TNFR-Related Gene
- HNSCC:
-
Head And Neck Squamous Cell Carcinoma
- ICT:
-
Immune Checkpoint Therapy
- TME:
-
Tumor Microenvironment
- MHC:
-
Major Histocompatibility Complex
- TCRs:
-
T Cell Receptors
- TILs:
-
Tumor-Infiltrating Lymphocytes
- MHC I:
-
Major Histocompatibility Complex Class I
- MSI:
-
Microsatellite Instability
- TMB:
-
Tumor Mutation Burden
- WES:
-
Whole Exome Sequencing
- MSI-H:
-
Microsatellite Instability High
- dMMR:
-
Mismatch Repair Deficiency
- pMMR:
-
Proficient Mismatch Repair
- DCs:
-
Dendritic Cells
- NK cells:
-
Natural Killer Cells
- CTLs:
-
Cytotoxic T Lymphocytes
- ORR:
-
Objective Response Rate
- CRC:
-
Colorectal Cancer
- RCC:
-
Renal Cell Carcinoma
- TNBC:
-
Triple-Negative Breast Cancer
- DAMPs:
-
Damage-Associated Molecular Patterns
- TAMs:
-
Tumor-Associated Macrophages
- ICD:
-
Immunogenic Cell Death
- SBRT:
-
Stereotactic Body Radiotherapy
- HDRT:
-
High-Dose Radiotherapy
- LDRT:
-
Low-Dose Radiotherapy
- CFRT:
-
Conventional Fractionated Radiotherapy
- MDSCs:
-
Myeloid-Derived Suppressor Cells
- GC:
-
Gastric Cancer
- CAR -T cells:
-
Chimeric Antigen Receptor T cells
- scFv:
-
single chain variable Fragment
- Ovs:
-
Oncolytic viruses
- T-VEC:
-
Talimogene Laherparepvec
- HSV-1:
-
Herpes Simplex Virus Type 1
- PTCV:
-
Personalized Therapeutic Cancer Vaccine
- CR:
-
Complete Response
- LOH:
-
Loss Of Heterozygosity
- β2m:
-
β2-microglobulin
- DNMT1:
-
DNA Methyltransferase 1
- DNMTis:
-
DNMT Inhibitors
- HDACis:
-
Histone Deacetylase Inhibitors
- Th1:
-
T-helper 1
- IRF1:
-
Interferon Regulatory Factor 1
- TCGA:
-
The Cancer Genome Atlas
- FMT:
-
Fecal Microbiota Transplantation
- EMT:
-
Epithelial-Mesenchymal Transition
- CAFs:
-
Cancer-Associated Fibroblasts
- FAP:
-
Fibroblast-Activating Protein
- iNOS:
-
inducible Nitric Oxide Synthase
- ROS:
-
Reactive Oxygen Species
- PDAC:
-
Pancreatic Ductal Adenocarcinoma
- OC:
-
Ovarian Cancer
- CPS:
-
Combined Positive Score
- TPS:
-
Tumor Proportion Score
- IC:
-
Immune Cell Score
- HERVs:
-
Human Endogenous Retroviruses
- dsRNA:
-
double-stranded RNA
- ITH:
-
Intratumor Heterogeneity
- CTCs:
-
Circulating Tumor Cells
- ctDNA:
-
circulating tumor DNA
- EVs:
-
Extracellular Vesicles
- AI:
-
Artificial Intelligence
- DLT:
-
Dose Limiting Toxicities
- TEAE:
-
Treatment-Emergent Adverse Event
- PFS:
-
Progression-Free Survival
- pCR:
-
pathological Complete Response
- RFS:
-
Recurrence-Free Survival
- DCR:
-
Disease Control Rate
- MPR:
-
Major Pathologic Response
- AEs:
-
Adverse Events
- HNSCC:
-
Head and Neck Squamous Cell Carcinoma
- ESCC:
-
Esophageal Squamous Cell Carcinoma
- GEJC:
-
Gastroesophageal Junction Cancer
- uHNSCC:
-
unresectable Head and Neck Squamous Cell Carcinoma
- DLBL:
-
Diffuse Large B Cell Lymphoma
- R/R NHL:
-
Relapsed/Refractory (R/R) Non-Hodgkin Lymphoma (NHL)
References
Khosravi GR, et al. Immunologic tumor microenvironment modulators for turning cold tumors hot. Cancer Commun (Lond). 2024;44(5):521–53.
Waldman AD, Fritz JM, Lenardo MJ. A guide to cancer immunotherapy: from T cell basic science to clinical practice. Nat Rev Immunol. 2020;20(11):651–68.
Chow A, et al. Clinical implications of T cell exhaustion for cancer immunotherapy. Nat Rev Clin Oncol. 2022;19(12):775–90.
Song E, Chow RD. Mutations in IFN-γ signaling genes sensitize tumors to immune checkpoint blockade. Cancer Cell. 2023;41(4):651–2.
Harris MA, et al. Towards targeting the breast cancer immune microenvironment. Nat Rev Cancer. 2024;24(8):554–77.
Pai JA, et al. Lineage tracing reveals clonal progenitors and long-term persistence of tumor-specific T cells during immune checkpoint blockade. Cancer Cell. 2023;41(4):776–e7907.
Blomberg OS, et al. IL-5-producing CD4(+) T cells and eosinophils cooperate to enhance response to immune checkpoint blockade in breast cancer. Cancer Cell. 2023;41(1):106–e12310.
van Elsas MJ, et al. Immunotherapy-activated T cells recruit and skew late-stage activated M1-like macrophages that are critical for therapeutic efficacy. Cancer Cell. 2024;42(6):1032–e105010.
Kaptein P, et al. CD8-Targeted IL2 unleashes tumor-specific immunity in Human Cancer tissue by reviving the dysfunctional T-cell Pool. Cancer Discov. 2024;14(7):1226–51.
Pai CS, et al. Clonal deletion of Tumor-Specific T cells by Interferon-γ confers therapeutic resistance to Combination Immune Checkpoint Blockade. Immunity. 2019;50(2):477–e4928.
Korman AJ, Garrett-Thomson SC, Lonberg N. The foundations of immune checkpoint blockade and the ipilimumab approval decennial. Nat Rev Drug Discov. 2022;21(7):509–28.
Rowshanravan B, Halliday N, Sansom DM. CTLA-4: a moving target in immunotherapy. Blood. 2018;131(1):58–67.
Robert C. A decade of immune-checkpoint inhibitors in cancer therapy. Nat Commun. 2020;11(1):3801.
Sharma P, Allison JP. Dissecting the mechanisms of immune checkpoint therapy. Nat Rev Immunol. 2020;20(2):75–6.
Sharma P, et al. Immune checkpoint therapy-current perspectives and future directions. Cell. 2023;186(8):1652–69.
Couzin-Frankel J. Breakthrough of the year 2013. Cancer Immunotherapy Sci. 2013;342(6165):1432–3.
Markham A, Dostarlimab. First Approval Drugs. 2021;81(10):1213–9.
Yarchoan M, et al. Personalized neoantigen vaccine and pembrolizumab in advanced hepatocellular carcinoma: a phase 1/2 trial. Nat Med. 2024;30(4):1044–53.
Mullard A. Second CTLA4-targeted checkpoint inhibitor secures FDA approval. Nat Rev Drug Discov. 2022;21(12):868.
Dhillon S, Penpulimab. First approval. Drugs. 2021;81(18):2159–66.
Hoy SM. Sintilimab: First Global approval. Drugs. 2019;79(3):341–6.
Markham A, Duggan S. Cemiplimab: First Global approval. Drugs. 2018;78(17):1841–6.
Syed YY, Durvalumab. First Global approval. Drugs. 2017;77(12):1369–76.
Marcus L, et al. Approval Summary: Pembrolizumab for the treatment of Tumor Mutational Burden-High Solid tumors. Clin Cancer Res. 2021;27(17):4685–9.
Cooper AJ, Yu HA. The Promise and limitations of Neoadjuvant Immune-Checkpoint Blockade in Resectable Non-small Cell Lung Cancer. Cancer Discov. 2023;13(11):2306–9.
Johnson PC, et al. Immune checkpoint inhibitors - the need for Innovation. N Engl J Med. 2023;388(16):1529–32.
Overman MJ et al. Nivolumab plus Relatlimab in patients with previously treated microsatellite instability-high/mismatch repair-deficient metastatic colorectal cancer: the phase II CheckMate 142 study. J Immunother Cancer, 2024. 12(5).
Cillo AR, 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(16):4373–e438815.
Hegewisch-Becker S, et al. First-line nivolumab and Relatlimab Plus Chemotherapy for gastric or gastroesophageal Junction Adenocarcinoma: the phase II RELATIVITY-060 study. J Clin Oncol. 2024;42(17):2080–93.
Park JS, et al. Targeting PD-L2-RGMb overcomes microbiome-related immunotherapy resistance. Nature. 2023;617(7960):377–85.
Bareche Y, et al. Leveraging big data of immune checkpoint blockade response identifies novel potential targets. Ann Oncol. 2022;33(12):1304–17.
Wong CW, et al. PARP14 inhibition restores PD-1 immune checkpoint inhibitor response following IFNγ-driven acquired resistance in preclinical cancer models. Nat Commun. 2023;14(1):5983.
Tawbi HA, et al. Relatlimab and Nivolumab versus Nivolumab in Untreated Advanced Melanoma. N Engl J Med. 2022;386(1):24–34.
Kelly RJ, et al. Neoadjuvant nivolumab or nivolumab plus LAG-3 inhibitor relatlimab in resectable esophageal/gastroesophageal junction cancer: a phase ib trial and ctDNA analyses. Nat Med. 2024;30(4):1023–34.
Schuler M, et al. Neoadjuvant nivolumab with or without relatlimab in resectable non-small-cell lung cancer: a randomized phase 2 trial. Nat Med. 2024;30(6):1602–11.
Bell HN, Zou W. Beyond the barrier: unraveling the mechanisms of Immunotherapy Resistance. Annu Rev Immunol. 2024;42(1):521–50.
Dall’Olio FG, et al. Tumour burden and efficacy of immune-checkpoint inhibitors. Nat Rev Clin Oncol. 2022;19(2):75–90.
Cai Z, et al. BCAT2 shapes a Noninflamed Tumor Microenvironment and induces resistance to Anti-PD-1/PD-L1 immunotherapy by negatively regulating Proinflammatory chemokines and Anticancer Immunity. Adv Sci (Weinh). 2023;10(8):e2207155.
Champiat S, Marabelle A. Paradoxical cancer cell stimulation by IFNγ drives tumor hyperprogression upon checkpoint blockade immunotherapy. Cancer Cell. 2023;41(2):229–31.
Guan X, et al. Androgen receptor activity in T cells limits checkpoint blockade efficacy. Nature. 2022;606(7915):791–6.
Attili I, Passaro A, de Marinis F. Anti-TIGIT to overcome resistance to immune checkpoint inhibitors in lung cancer: limits and potentials. Ann Oncol. 2022;33(2):119–22.
Powles T, et al. Atezolizumab with enzalutamide versus enzalutamide alone in metastatic castration-resistant prostate cancer: a randomized phase 3 trial. Nat Med. 2022;28(1):144–53.
Butterfield LH, Najjar YG. Immunotherapy combination approaches: mechanisms, biomarkers and clinical observations. Nat Rev Immunol. 2024;24(6):399–416.
Marangoni F, et al. Expansion of tumor-associated Treg cells upon disruption of a CTLA-4-dependent feedback loop. Cell. 2021;184(15):3998–e401519.
O’Malley DM, et al. Dual PD-1 and CTLA-4 checkpoint blockade using Balstilimab and Zalifrelimab Combination as Second-Line treatment for Advanced Cervical Cancer: an open-label phase II study. J Clin Oncol. 2022;40(7):762–71.
de Miguel M, Calvo E. Clinical challenges of Immune Checkpoint inhibitors. Cancer Cell. 2020;38(3):326–33.
Zugazagoitia J, Paz-Ares L. Extensive-stage small-cell Lung Cancer: first-line and second-line treatment options. J Clin Oncol. 2022;40(6):671–80.
Chong X, et al. Recent developments in immunotherapy for gastrointestinal tract cancers. J Hematol Oncol. 2024;17(1):65.
Holder AM, et al. Defining clinically useful biomarkers of immune checkpoint inhibitors in solid tumours. Nat Rev Cancer. 2024;24(7):498–512.
Haddad AF, et al. Resistance to immune checkpoint blockade: Mechanisms, counter-acting approaches, and future directions. Semin Cancer Biol. 2022;86(Pt 3):532–41.
Gutierrez M, et al. Biomarker-directed, pembrolizumab-based combination therapy in non-small cell lung cancer: phase 2 KEYNOTE-495/KeyImPaCT trial interim results. Nat Med. 2023;29(7):1718–27.
Jardim DL, et al. The challenges of Tumor Mutational Burden as an Immunotherapy Biomarker. Cancer Cell. 2021;39(2):154–73.
Smithy JW, Luke JJ. An emerging biomarker for combined CTLA-4 and PD-1 blockade. Clin Cancer Res. 2023;29(13):2345–7.
Nishino M, et al. Monitoring immune-checkpoint blockade: response evaluation and biomarker development. Nat Rev Clin Oncol. 2017;14(11):655–68.
Besse B, et al. Biomarker-directed targeted therapy plus durvalumab in advanced non-small-cell lung cancer: a phase 2 umbrella trial. Nat Med. 2024;30(3):716–29.
Pérez-Ruiz E, et al. Cancer immunotherapy resistance based on immune checkpoints inhibitors: targets, biomarkers, and remedies. Drug Resist Updat. 2020;53:100718.
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;16:223–49.
Fan Z, et al. Blocking interaction between SHP2 and PD-1 denotes a novel opportunity for developing PD-1 inhibitors. EMBO Mol Med. 2020;12(6):e11571.
Hebeisen M, et al. SHP-1 phosphatase activity counteracts increased T cell receptor affinity. J Clin Invest. 2013;123(3):1044–56.
Hsu LY, et al. Destabilizing the autoinhibitory conformation of Zap70 induces up-regulation of inhibitory receptors and T cell unresponsiveness. J Exp Med. 2017;214(3):833–49.
Larsen TV, Hussmann D, Nielsen AL. PD-L1 and PD-L2 expression correlated genes in non-small-cell lung cancer. Cancer Commun (Lond). 2019;39(1):30.
Hurrell BP, et al. PD-L2 controls peripherally induced regulatory T cells by maintaining metabolic activity and Foxp3 stability. Nat Commun. 2022;13(1):5118.
Saliakoura M, et al. PLCγ1 suppression promotes the adaptation of KRAS-mutant lung adenocarcinomas to hypoxia. Nat Cell Biol. 2020;22(11):1382–95.
Noh DY, et al. Elevated content of phospholipase C-gamma 1 in colorectal cancer tissues. Cancer. 1994;73(1):36–41.
Zhu Z, et al. Improving cancer immunotherapy by rationally combining oncolytic virus with modulators targeting key signaling pathways. Mol Cancer. 2022;21(1):196.
Kumagai S, et al. An oncogenic alteration creates a Microenvironment that promotes Tumor Progression by conferring a Metabolic Advantage to Regulatory T Cells. Immunity. 2020;53(1):187–e2038.
Wu RC, et al. Identification of the PTEN-ARID4B-PI3K pathway reveals the dependency on ARID4B by PTEN-deficient prostate cancer. Nat Commun. 2019;10(1):4332.
Mukherjee R, et al. Regulation of PTEN translation by PI3K signaling maintains pathway homeostasis. Mol Cell. 2021;81(4):708–e7235.
Chida K, et al. A low tumor mutational burden and PTEN mutations are predictors of a negative response to PD-1 blockade in MSI-H/dMMR gastrointestinal tumors. Clin Cancer Res. 2021;27(13):3714–24.
Ferguson R, et al. Germline immunomodulatory expression quantitative trait loci (ieQTLs) associated with immune-related toxicity from checkpoint inhibition. Eur J Cancer. 2023;189:112923.
Liu M, et al. Myeloid-derived suppressor cells regulate the immunosuppressive functions of PD-1(-)PD-L1(+) bregs through PD-L1/PI3K/AKT/NF-κB axis in breast cancer. Cell Death Dis. 2021;12(5):465.
Sznol M, Melero I. Revisiting anti-CTLA-4 antibodies in combination with PD-1 blockade for cancer immunotherapy. Ann Oncol. 2021;32(3):295–7.
Burke KP, et al. The B7:CD28 family and friends: unraveling coinhibitory interactions. Immunity. 2024;57(2):223–44.
Guo J, Xue Z, Wang L. Transcriptional regulation of the immune checkpoints PD-1 and CTLA-4. Cell Mol Immunol. 2022;19(7):861–2.
Gaikwad S, et al. Immune checkpoint proteins: signaling mechanisms and molecular interactions in cancer immunotherapy. Semin Cancer Biol. 2022;86(Pt 3):137–50.
Wei SC, et al. Combination anti-CTLA-4 plus anti-PD-1 checkpoint blockade utilizes cellular mechanisms partially distinct from monotherapies. Proc Natl Acad Sci U S A. 2019;116(45):22699–709.
Dovedi SJ, et al. Design and efficacy of a Monovalent Bispecific PD-1/CTLA4 antibody that enhances CTLA4 blockade on PD-1(+) activated T cells. Cancer Discov. 2021;11(5):1100–17.
Campbell KM, et al. Prior anti-CTLA-4 therapy impacts molecular characteristics associated with anti-PD-1 response in advanced melanoma. Cancer Cell. 2023;41(4):791–e8064.
Robert C, et al. Pembrolizumab versus Ipilimumab in Advanced Melanoma. N Engl J Med. 2015;372(26):2521–32.
Schachter J, 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(10105):1853–62.
Robert C, et al. Pembrolizumab versus Ipilimumab in advanced melanoma (KEYNOTE-006): post-hoc 5-year results from an open-label, multicentre, randomised, controlled, phase 3 study. Lancet Oncol. 2019;20(9):1239–51.
Poole RM. Pembrolizumab: first global approval. Drugs. 2014;74(16):1973–81.
Brahmer JR, et al. Phase I study of single-agent anti-programmed death-1 (MDX-1106) in refractory solid tumors: safety, clinical activity, pharmacodynamics, and immunologic correlates. J Clin Oncol. 2010;28(19):3167–75.
Larkin J, et al. Combined Nivolumab and Ipilimumab or Monotherapy in untreated melanoma. N Engl J Med. 2015;373(1):23–34.
Robert C, et al. Nivolumab in previously untreated melanoma without BRAF mutation. N Engl J Med. 2015;372(4):320–30.
Reck M, et al. Pembrolizumab versus Chemotherapy for PD-L1-Positive non-small-cell Lung Cancer. N Engl J Med. 2016;375(19):1823–33.
Antonia SJ, et al. Durvalumab after Chemoradiotherapy in Stage III Non-small-cell Lung Cancer. N Engl J Med. 2017;377(20):1919–29.
Horn L, et al. First-line atezolizumab plus chemotherapy in extensive-stage small-cell Lung Cancer. N Engl J Med. 2018;379(23):2220–9.
Paz-Ares L, 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(10212):1929–39.
Powles T, et al. Avelumab Maintenance Therapy for Advanced or Metastatic Urothelial Carcinoma. N Engl J Med. 2020;383(13):1218–30.
Le DT, et al. PD-1 blockade in tumors with Mismatch-Repair Deficiency. N Engl J Med. 2015;372(26):2509–20.
Chambers CA, et al. CTLA-4-mediated inhibition in regulation of T cell responses: mechanisms and manipulation in tumor immunotherapy. Annu Rev Immunol. 2001;19:565–94.
Leach DR, Krummel MF, Allison JP. Enhancement of antitumor immunity by CTLA-4 blockade. Science. 1996;271(5256):1734–6.
Waterhouse P, et al. Lymphoproliferative disorders with early lethality in mice deficient in Ctla-4. Science. 1995;270(5238):985–8.
Walunas TL, et al. CTLA-4 can function as a negative regulator of T cell activation. Immunity. 1994;1(5):405–13.
Xu X et al. CTLA4 depletes T cell endogenous and trogocytosed B7 ligands via cis-endocytosis. J Exp Med, 2023. 220(7).
Kaptein P, et al. Addition of interleukin-2 overcomes resistance to neoadjuvant CTLA4 and PD1 blockade in ex vivo patient tumors. Sci Transl Med. 2022;14(642):eabj9779.
Wang CJ, et al. Costimulation blockade in combination with IL-2 permits regulatory T cell sparing immunomodulation that inhibits autoimmunity. Nat Commun. 2022;13(1):6757.
Yamaguchi T, et al. Construction of self-recognizing regulatory T cells from conventional T cells by controlling CTLA-4 and IL-2 expression. Proc Natl Acad Sci U S A. 2013;110(23):E2116–25.
Krummel MF, Allison JP. CTLA-4 engagement inhibits IL-2 accumulation and cell cycle progression upon activation of resting T cells. J Exp Med. 1996;183(6):2533–40.
Chuang E, et al. The CD28 and CTLA-4 receptors associate with the serine/threonine phosphatase PP2A. Immunity. 2000;13(3):313–22.
Chand D et al. Botensilimab, an Fc-enhanced anti-CTLA-4 antibody, is effective against tumors poorly responsive to conventional immunotherapy. Cancer Discov, 2024.
Frijlink E et al. PD-1 or CTLA-4 blockade promotes CD86-driven Treg responses upon radiotherapy of lymphocyte-depleted cancer in mice. J Clin Invest, 2024. 134(6).
Goswami S, et al. Modulation of EZH2 expression in T cells improves efficacy of anti-CTLA-4 therapy. J Clin Invest. 2018;128(9):3813–8.
Keam SJ, Tremelimumab. First Approval Drugs. 2023;83(1):93–102.
Hodi FS, et al. Improved survival with ipilimumab in patients with metastatic melanoma. N Engl J Med. 2010;363(8):711–23.
VanderWalde A, et al. Ipilimumab with or without nivolumab in PD-1 or PD-L1 blockade refractory metastatic melanoma: a randomized phase 2 trial. Nat Med. 2023;29(9):2278–85.
Zamarin D, et al. Randomized phase II trial of Nivolumab Versus Nivolumab and Ipilimumab for recurrent or persistent ovarian Cancer: an NRG Oncology Study. J Clin Oncol. 2020;38(16):1814–23.
Slovin SF, et al. Ipilimumab alone or in combination with radiotherapy in metastatic castration-resistant prostate cancer: results from an open-label, multicenter phase I/II study. Ann Oncol. 2013;24(7):1813–21.
Beer TM, et al. Randomized, Double-Blind, phase III trial of Ipilimumab Versus Placebo in Asymptomatic or minimally symptomatic patients with metastatic chemotherapy-naive castration-resistant prostate Cancer. J Clin Oncol. 2017;35(1):40–7.
Small EJ, et al. A pilot trial of CTLA-4 blockade with human anti-CTLA-4 in patients with hormone-refractory prostate cancer. Clin Cancer Res. 2007;13(6):1810–5.
Weber JS, et al. Phase I/II study of ipilimumab for patients with metastatic melanoma. J Clin Oncol. 2008;26(36):5950–6.
Phan GQ, et al. Cancer regression and autoimmunity induced by cytotoxic T lymphocyte-associated antigen 4 blockade in patients with metastatic melanoma. Proc Natl Acad Sci U S A. 2003;100(14):8372–7.
Ledford H. Melanoma drug wins US approval. Nature. 2011;471(7340):561.
Ribas A, et al. Phase III randomized clinical trial comparing tremelimumab with standard-of-care chemotherapy in patients with advanced melanoma. J Clin Oncol. 2013;31(5):616–22.
Kelley RK, et al. Safety, Efficacy, and Pharmacodynamics of Tremelimumab Plus Durvalumab for patients with Unresectable Hepatocellular Carcinoma: Randomized Expansion of a phase I/II study. J Clin Oncol. 2021;39(27):2991–3001.
Kelley RK et al. Efficacy, tolerability, and biologic activity of a novel regimen of tremelimumab (T) in combination with durvalumab (D) for patients (pts) with advanced hepatocellular carcinoma (aHCC). 2020. 38(15_suppl): pp. 4508–4508.
Abou-Alfa GK, et al. Tremelimumab plus Durvalumab in Unresectable Hepatocellular Carcinoma. NEJM Evid. 2022;1(8):EVIDoa2100070.
Johnson ML, 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(6):1213–27.
!!!. INVALID CITATION !!! [95–100].
Cai L, et al. Targeting LAG-3, TIM-3, and TIGIT for cancer immunotherapy. J Hematol Oncol. 2023;16(1):101.
Qin S, et al. Novel immune checkpoint targets: moving beyond PD-1 and CTLA-4. Mol Cancer. 2019;18(1):155.
Joller N, Anderson AC, Kuchroo VK. LAG-3, TIM-3, and TIGIT: distinct functions in immune regulation. Immunity. 2024;57(2):206–22.
Maruhashi T, et al. Binding of LAG-3 to stable peptide-MHC class II limits T cell function and suppresses autoimmunity and anti-cancer immunity. Immunity. 2022;55(5):912–e9248.
Andrews LP, et al. LAG-3 and PD-1 synergize on CD8(+) T cells to drive T cell exhaustion and hinder autocrine IFN-γ-dependent anti-tumor immunity. Cell. 2024;187(16):4355–e437222.
Adkins D et al. TACTI-003: a randomized phase IIb study of eftilagimod alpha (soluble LAG-3 protein) and pembrolizumab as first-line treatment of patients with recurrent or metastatic head and neck squamous cell carcinoma. 2022. 40(16_suppl): p. TPS6099–6099.
Liu F, Liu Y, Chen Z. Tim-3 expression and its role in hepatocellular carcinoma. J Hematol Oncol. 2018;11(1):126.
Lu L, Deng L. TIM-3 inhibitors: a promising strategy for tumor immunotherapy. Trends Mol Med. 2024;30(3):202–3.
Pagliano O et al. Tim-3 mediates T cell trogocytosis to limit antitumor immunity. J Clin Invest, 2022. 132(9).
Yang R, et al. Galectin-9 interacts with PD-1 and TIM-3 to regulate T cell death and is a target for cancer immunotherapy. Nat Commun. 2021;12(1):832.
Curigliano G, et al. Phase I/Ib clinical trial of Sabatolimab, an Anti-TIM-3 antibody, alone and in combination with Spartalizumab, an Anti-PD-1 antibody, in Advanced Solid tumors. Clin Cancer Res. 2021;27(13):3620–9.
Zeidan AM, et al. Sabatolimab plus hypomethylating agents in previously untreated patients with higher-risk myelodysplastic syndromes (STIMULUS-MDS1): a randomised, double-blind, placebo-controlled, phase 2 trial. Lancet Haematol. 2024;11(1):e38–50.
Dama P, et al. Gal9/Tim-3 expression level is higher in AML patients who fail chemotherapy. J Immunother Cancer. 2019;7(1):175.
Brunner AM, et al. Phase ib study of sabatolimab (MBG453), a novel immunotherapy targeting TIM-3 antibody, in combination with decitabine or azacitidine in high- or very high-risk myelodysplastic syndromes. Am J Hematol. 2024;99(2):E32–6.
Hollebecque A, et al. Safety and Antitumor Activity of α-PD-L1 antibody as Monotherapy or in combination with α-TIM-3 antibody in patients with microsatellite Instability-High/Mismatch repair-deficient tumors. Clin Cancer Res. 2021;27(23):6393–404.
Brunner AM, et al. Efficacy and safety of Sabatolimab (MBG453) in combination with Hypomethylating agents (HMAs) in patients (pts) with very High/High-Risk myelodysplastic syndrome (vHR/HR-MDS) and Acute Myeloid Leukemia (AML): final analysis from a phase ib study. Blood. 2021;138(Supplement 1):244–244.
Falchook GS et al. Phase 1 trial of TIM-3 inhibitor cobolimab monotherapy and in combination with PD-1 inhibitors nivolumab or dostarlimab (AMBER). 2022. 40(16_suppl): pp. 2504–2504.
Niu J, 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(2):169–80.
Cho BC, et al. Tiragolumab plus Atezolizumab versus placebo plus atezolizumab as a first-line treatment for PD-L1-selected non-small-cell lung cancer (CITYSCAPE): primary and follow-up analyses of a randomised, double-blind, phase 2 study. Lancet Oncol. 2022;23(6):781–92.
Frentzas S et al. AdvanTIG-105: a phase I dose escalation study of the anti-TIGIT monoclonal antibody ociperlimab in combination with tislelizumab in patients with advanced solid tumors. J Immunother Cancer, 2023. 11(10).
Mettu NB, et al. A phase 1a/b Open-Label, dose-escalation study of Etigilimab Alone or in combination with Nivolumab in patients with locally advanced or metastatic solid tumors. Clin Cancer Res. 2022;28(5):882–92.
Finn RS et al. Results from the MORPHEUS-liver study: phase Ib/II randomized evaluation of tiragolumab (tira) in combination with atezolizumab (atezo) and bevacizumab (bev) in patients with unresectable, locally advanced or metastatic hepatocellular carcinoma (uHCC). 2023. 41(16_suppl): pp. 4010–4010.
Rudin CM, SKYSCRAPER-02. : Primary results of a phase III, randomized, double-blind, placebo-controlled study of atezolizumab (atezo) + carboplatin + etoposide (CE) with or without tiragolumab (tira) in patients (pts) with untreated extensive-stage small cell lung cancer (ES-SCLC). 2022. 40(17_suppl): p. LBA8507–8507.
Peters S, et al. P14.03 Vibostolimab Plus Pembrolizumab With/Without Docetaxel vs Docetaxel in NSCLC after Platinum Chemotherapy and Immunotherapy. J Thorac Oncol. 2021;16(10, Supplement):pS1011–S1012.
Burris HA et al. Phase 1 safety of ICOS agonist antibody JTX-2011 alone and with nivolumab (nivo) in advanced solid tumors; predicted vs observed pharmacokinetics (PK) in ICONIC. 2017. 35(15_suppl): pp. 3033–3033.
Kobziev O et al. Phase 2 study of PD-1 inhibitor JTX-4014 alone and in combination with vopratelimab, an ICOS agonist, in biomarker-selected subjects with metastatic NSCLC after one prior platinum-containing regimen (SELECT). 2021. 39(15_suppl): p. TPS9137–9137.
Ávila Ávila A, et al. Targeting the TNF/IAP pathway synergizes with anti-CD3 immunotherapy in T-cell acute lymphoblastic leukemia. Blood. 2024;143(21):2166–77.
An Anti-PD-1-GITR Ligand Bispecific Enhances Antitumor Immunity. Cancer Discov, 2022. 12(5): p. Of10.
Yu X, et al. Reducing affinity as a strategy to boost immunomodulatory antibody agonism. Nature. 2023;614(7948):539–47.
Hirschhorn D, et al. T cell immunotherapies engage neutrophils to eliminate tumor antigen escape variants. Cell. 2023;186(7):1432–e144717.
Cappell KM, Kochenderfer JN. A comparison of chimeric antigen receptors containing CD28 versus 4-1BB costimulatory domains. Nat Rev Clin Oncol. 2021;18(11):715–27.
Kraehenbuehl L, et al. Enhancing immunotherapy in cancer by targeting emerging immunomodulatory pathways. Nat Rev Clin Oncol. 2022;19(1):37–50.
Duhen R, et al. Neoadjuvant anti-OX40 (MEDI6469) therapy in patients with head and neck squamous cell carcinoma activates and expands antigen-specific tumor-infiltrating T cells. Nat Commun. 2021;12(1):1047.
Gutierrez M, et al. OX40 agonist BMS-986178 alone or in Combination with Nivolumab and/or ipilimumab in patients with Advanced Solid tumors. Clin Cancer Res. 2021;27(2):460–72.
Kim TW, et al. First-In-Human phase I study of the OX40 agonist MOXR0916 in patients with Advanced Solid tumors. Clin Cancer Res. 2022;28(16):3452–63.
Balmanoukian AS, et al. Safety and clinical activity of MEDI1873, a Novel GITR Agonist, in Advanced Solid tumors. Clin Cancer Res. 2020;26(23):6196–203.
Papadopoulos KP, et al. Phase I study of MK-4166, an anti-human glucocorticoid-Induced TNF receptor antibody, alone or with Pembrolizumab in Advanced Solid tumors. Clin Cancer Res. 2021;27(7):1904–11.
Piha-Paul SA et al. First-in-human phase I/Ib open-label dose-escalation study of GWN323 (anti-GITR) as a single agent and in combination with spartalizumab (anti-PD-1) in patients with advanced solid tumors and lymphomas. J Immunother Cancer, 2021. 9(8).
Scott EC, et al. Trends in the approval of cancer therapies by the FDA in the twenty-first century. Nat Rev Drug Discov. 2023;22(8):625–40.
Subbiah V et al. The evolving landscape of tissue-agnostic therapies in precision oncology. CA Cancer J Clin, 2024.
Gong J, et al. Combination systemic therapies with immune checkpoint inhibitors in pancreatic cancer: overcoming resistance to single-agent checkpoint blockade. Clin Transl Med. 2018;7(1):32.
Nassiri F, et al. Oncolytic DNX-2401 virotherapy plus pembrolizumab in recurrent glioblastoma: a phase 1/2 trial. Nat Med. 2023;29(6):1370–8.
Clinton TN. Defining resistance to Immune checkpoint inhibitors in Urothelial Carcinoma: insights from PURE-01. Eur Urol. 2021;80(3):323–4.
Gide TN, et al. Primary and Acquired Resistance to Immune checkpoint inhibitors in metastatic melanoma. Clin Cancer Res. 2018;24(6):1260–70.
Kim JM, Chen DS. Immune escape to PD-L1/PD-1 blockade: seven steps to success (or failure). Ann Oncol. 2016;27(8):1492–504.
Chen DS, Mellman I. Elements of cancer immunity and the cancer-immune set point. Nature. 2017;541(7637):321–30.
Huang Q, et al. The primordial differentiation of tumor-specific memory CD8(+) T cells as bona fide responders to PD-1/PD-L1 blockade in draining lymph nodes. Cell. 2022;185(22):4049–e406625.
Vignali PDA, et al. Hypoxia drives CD39-dependent suppressor function in exhausted T cells to limit antitumor immunity. Nat Immunol. 2023;24(2):267–79.
Kersten K, et al. Spatiotemporal co-dependency between macrophages and exhausted CD8(+) T cells in cancer. Cancer Cell. 2022;40(6):624–e6389.
Galon J, Bruni D. Approaches to treat immune hot, altered and cold tumours with combination immunotherapies. Nat Rev Drug Discov. 2019;18(3):197–218.
Bruni D, Angell HK, Galon J. The immune contexture and immunoscore in cancer prognosis and therapeutic efficacy. Nat Rev Cancer. 2020;20(11):662–80.
Yang Y, et al. Tislelizumab plus chemotherapy as first-line treatment for recurrent or metastatic nasopharyngeal cancer: a multicenter phase 3 trial (RATIONALE-309). Cancer Cell. 2023;41(6):1061–e10724.
Enfield KSS, et al. Spatial Architecture of myeloid and T cells orchestrates Immune Evasion and Clinical Outcome in Lung Cancer. Cancer Discov. 2024;14(6):1018–47.
Doroshow DB, et al. PD-L1 as a biomarker of response to immune-checkpoint inhibitors. Nat Rev Clin Oncol. 2021;18(6):345–62.
Ott PA, 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(4):318–27.
Gibney GT, Weiner LM, Atkins MB. Predictive biomarkers for checkpoint inhibitor-based immunotherapy. Lancet Oncol. 2016;17(12):e542–51.
Mountzios G, et al. Position of an international panel of lung cancer experts on the decision for expansion of approval for pembrolizumab in advanced non-small-cell lung cancer with a PD-L1 expression level of ≥ 1% by the USA Food and Drug Administration. Ann Oncol. 2019;30(11):1686–8.
Davis AA, Patel VG. The role of PD-L1 expression as a predictive biomarker: an analysis of all US Food and Drug Administration (FDA) approvals of immune checkpoint inhibitors. J Immunother Cancer. 2019;7(1):278.
Wu HX, et al. Clinical benefit of first-line programmed Death-1 antibody plus chemotherapy in low programmed cell death ligand 1-Expressing esophageal squamous cell carcinoma: a Post Hoc Analysis of JUPITER-06 and Meta-analysis. J Clin Oncol. 2023;41(9):1735–46.
Zviran A, et al. Genome-wide cell-free DNA mutational integration enables ultra-sensitive cancer monitoring. Nat Med. 2020;26(7):1114–24.
Wang X, et al. Tumor mutational burden for the prediction of PD-(L)1 blockade efficacy in cancer: challenges and opportunities. Ann Oncol. 2024;35(6):508–22.
Thein KZ, Lemery SJ, Kummar S. Tissue-agnostic Drug Development: a new path to drug approval. Cancer Discov. 2021;11(9):2139–44.
Nassar AH, et al. Ancestry-driven recalibration of tumor mutational burden and disparate clinical outcomes in response to immune checkpoint inhibitors. Cancer Cell. 2022;40(10):1161–e11725.
Valero C, et al. Response rates to Anti-PD-1 immunotherapy in microsatellite-stable solid tumors with 10 or more mutations per megabase. JAMA Oncol. 2021;7(5):739–43.
Fang LT, et al. Establishing community reference samples, data and call sets for benchmarking cancer mutation detection using whole-genome sequencing. Nat Biotechnol. 2021;39(9):1151–60.
Stenzinger A, et al. Harmonization and standardization of panel-based Tumor Mutational Burden Measurement: real-world results and recommendations of the quality in Pathology Study. J Thorac Oncol. 2020;15(7):1177–89.
Luchini C, et al. ESMO recommendations on microsatellite instability testing for immunotherapy in cancer, and its relationship with PD-1/PD-L1 expression and tumour mutational burden: a systematic review-based approach. Ann Oncol. 2019;30(8):1232–43.
Wang Z, et al. Comutations in DNA damage response pathways serve as potential biomarkers for Immune Checkpoint Blockade. Cancer Res. 2018;78(22):6486–96.
Asaoka Y, Ijichi H, Koike K. PD-1 blockade in tumors with Mismatch-Repair Deficiency. N Engl J Med. 2015;373(20):1979.
Bhamidipati D, Subbiah V. Tumor-agnostic drug development in dMMR/MSI-H solid tumors. Trends Cancer. 2023;9(10):828–39.
Robertson BM, et al. Determinants of resistance and response to melanoma therapy. Nat Cancer. 2024;5(7):964–82.
Yi M, et al. Combination strategies with PD-1/PD-L1 blockade: current advances and future directions. Mol Cancer. 2022;21(1):28.
Wolchok JD, et al. Long-term outcomes with Nivolumab Plus Ipilimumab or Nivolumab alone Versus Ipilimumab in patients with Advanced Melanoma. J Clin Oncol. 2022;40(2):127–37.
Tawbi HA, et al. Long-term outcomes of patients with active melanoma brain metastases treated with combination nivolumab plus ipilimumab (CheckMate 204): final results of an open-label, multicentre, phase 2 study. Lancet Oncol. 2021;22(12):1692–704.
Brahmer JR, et al. Five-year survival outcomes with Nivolumab Plus Ipilimumab Versus Chemotherapy as First-Line treatment for metastatic non-small-cell lung Cancer in CheckMate 227. J Clin Oncol. 2023;41(6):1200–12.
Lenz HJ, 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(2):161–70.
Chalabi M, et al. Neoadjuvant immunotherapy leads to pathological responses in MMR-proficient and MMR-deficient early-stage colon cancers. Nat Med. 2020;26(4):566–76.
Rudin CM, et al. SKYSCRAPER-02: Tiragolumab in Combination with Atezolizumab Plus Chemotherapy in untreated extensive-stage small-cell Lung Cancer. J Clin Oncol. 2024;42(3):324–35.
Pomeroy AE, et al. Drug independence and the curability of cancer by combination chemotherapy. Trends Cancer. 2022;8(11):915–29.
Galluzzi L, et al. Immunostimulation with chemotherapy in the era of immune checkpoint inhibitors. Nat Rev Clin Oncol. 2020;17(12):725–41.
Zhou C, et al. A global phase 3 study of serplulimab plus chemotherapy as first-line treatment for advanced squamous non-small-cell lung cancer (ASTRUM-004). Cancer Cell. 2024;42(2):198–e2083.
Cheng Y, et al. Effect of First-Line Serplulimab vs Placebo added to Chemotherapy on Survival in patients with extensive-stage small cell Lung Cancer: the ASTRUM-005 Randomized Clinical Trial. JAMA. 2022;328(12):1223–32.
Liu SV, et al. Updated overall survival and PD-L1 subgroup analysis of patients with extensive-stage small-cell lung Cancer treated with atezolizumab, carboplatin, and Etoposide (IMpower133). J Clin Oncol. 2021;39(6):619–30.
Mittendorf EA, et al. Neoadjuvant atezolizumab in combination with sequential nab-paclitaxel and anthracycline-based chemotherapy versus placebo and chemotherapy in patients with early-stage triple-negative breast cancer (IMpassion031): a randomised, double-blind, phase 3 trial. Lancet. 2020;396(10257):1090–100.
Janjigian YY, 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(10294):27–40.
Oh DY, et al. Durvalumab plus Gemcitabine and Cisplatin in Advanced biliary tract Cancer. NEJM Evid. 2022;1(8):EVIDoa2200015.
Zhang Z, et al. Involvement of inflammasomes in tumor microenvironment and tumor therapies. J Hematol Oncol. 2023;16(1):24.
Kim R, et al. Early Tumor-Immune Microenvironmental remodeling and response to First-Line Fluoropyrimidine and Platinum Chemotherapy in Advanced Gastric Cancer. Cancer Discov. 2022;12(4):984–1001.
Zhu S, et al. Combination strategies to maximize the benefits of cancer immunotherapy. J Hematol Oncol. 2021;14(1):156.
Forde PM, et al. Neoadjuvant Nivolumab plus Chemotherapy in Resectable Lung Cancer. N Engl J Med. 2022;386(21):1973–85.
Deutsch JS, et al. Association between pathologic response and survival after neoadjuvant therapy in lung cancer. Nat Med. 2024;30(1):218–28.
Cortes J, et al. 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(10265):1817–28.
Kang YK, et al. Nivolumab plus chemotherapy versus placebo plus chemotherapy in patients with HER2-negative, untreated, unresectable advanced or recurrent gastric or gastro-oesophageal junction cancer (ATTRACTION-4): a randomised, multicentre, double-blind, placebo-controlled, phase 3 trial. Lancet Oncol. 2022;23(2):234–47.
Akinboro O, Food US, Drug Administration Approval Summary. Nivolumab Plus Platinum-Doublet Chemotherapy for the Neoadjuvant treatment of patients with Resectable Non-small-cell Lung Cancer. J Clin Oncol. 2023;41(17):3249–59.
Motzer RJ, et al. Adjuvant nivolumab plus ipilimumab versus placebo for localised renal cell carcinoma after nephrectomy (CheckMate 914): a double-blind, randomised, phase 3 trial. Lancet. 2023;401(10379):821–32.
Oaknin A, et al. Nivolumab with or without ipilimumab in patients with recurrent or metastatic cervical cancer (CheckMate 358): a phase 1–2, open-label, multicohort trial. Lancet Oncol. 2024;25(5):588–602.
Provencio M, et al. Perioperative Nivolumab and Chemotherapy in Stage III Non-small-cell Lung Cancer. N Engl J Med. 2023;389(6):504–13.
Skrede OJ, et al. Deep learning for prediction of colorectal cancer outcome: a discovery and validation study. Lancet. 2020;395(10221):350–60.
Lee SM, et al. First-line atezolizumab monotherapy versus single-agent chemotherapy in patients with non-small-cell lung cancer ineligible for treatment with a platinum-containing regimen (IPSOS): a phase 3, global, multicentre, open-label, randomised controlled study. Lancet. 2023;402(10400):451–63.
Loibl S, et al. A randomised phase II study investigating durvalumab in addition to an anthracycline taxane-based neoadjuvant therapy in early triple-negative breast cancer: clinical results and biomarker analysis of GeparNuevo study. Ann Oncol. 2019;30(8):1279–88.
Fernandez-Martinez A, et al. Prognostic and predictive value of Immune-related gene expression signatures vs Tumor-infiltrating lymphocytes in early-stage ERBB2/HER2-Positive breast Cancer: a correlative analysis of the CALGB 40601 and PAMELA Trials. JAMA Oncol. 2023;9(4):490–9.
McLaughlin M, et al. Inflammatory microenvironment remodelling by tumour cells after radiotherapy. Nat Rev Cancer. 2020;20(4):203–17.
Oyoshi H, et al. Comprehensive single-cell analysis demonstrates radiotherapy-induced infiltration of macrophages expressing immunosuppressive genes into tumor in esophageal squamous cell carcinoma. Sci Adv. 2023;9(50):eadh9069.
Hou Y, et al. Radiotherapy enhances Metastasis through Immune suppression by inducing PD-L1 and MDSC in Distal sites. Clin Cancer Res. 2024;30(9):1945–58.
Hui R, et al. Patient-reported outcomes with durvalumab after chemoradiotherapy in stage III, unresectable non-small-cell lung cancer (PACIFIC): a randomised, controlled, phase 3 study. Lancet Oncol. 2019;20(12):1670–80.
Spigel DR, et al. Five-year survival outcomes from the PACIFIC Trial: Durvalumab after Chemoradiotherapy in Stage III Non-small-cell Lung Cancer. J Clin Oncol. 2022;40(12):1301–11.
Garassino MC, et al. Durvalumab after Sequential Chemoradiotherapy in Stage III, Unresectable NSCLC: the phase 2 PACIFIC-6 Trial. J Thorac Oncol. 2022;17(12):1415–27.
Altorki NK, et al. Neoadjuvant durvalumab with or without stereotactic body radiotherapy in patients with early-stage non-small-cell lung cancer: a single-centre, randomised phase 2 trial. Lancet Oncol. 2021;22(6):824–35.
Theelen W, et al. Pembrolizumab with or without radiotherapy for metastatic non-small-cell lung cancer: a pooled analysis of two randomised trials. Lancet Respir Med. 2021;9(5):467–75.
Theelen W, et al. Effect of Pembrolizumab after stereotactic body radiotherapy vs Pembrolizumab alone on Tumor response in patients with Advanced Non-small Cell Lung Cancer: results of the PEMBRO-RT phase 2 Randomized Clinical Trial. JAMA Oncol. 2019;5(9):1276–82.
Klug F, et al. Low-dose irradiation programs macrophage differentiation to an iNOS⁺/M1 phenotype that orchestrates effective T cell immunotherapy. Cancer Cell. 2013;24(5):589–602.
Herrera FG, et al. Low-dose Radiotherapy reverses Tumor Immune Desertification and Resistance to Immunotherapy. Cancer Discov. 2022;12(1):108–33.
Schoenfeld JD, et al. Durvalumab plus Tremelimumab alone or in combination with low-dose or hypofractionated radiotherapy in metastatic non-small-cell lung cancer refractory to previous PD(L)-1 therapy: an open-label, multicentre, randomised, phase 2 trial. Lancet Oncol. 2022;23(2):279–91.
Patel RR, et al. High-dose irradiation in combination with non-ablative low-dose radiation to treat metastatic disease after progression on immunotherapy: results of a phase II trial. Radiother Oncol. 2021;162:60–7.
Wang H et al. Preclinical study and phase II trial of adapting low-dose radiotherapy to immunotherapy in small cell lung cancer. Med, 2024.
Patel RB et al. Low-dose targeted radionuclide therapy renders immunologically cold tumors responsive to immune checkpoint blockade. Sci Transl Med, 2021. 13(602).
Li S, et al. Low-dose radiotherapy combined with dual PD-L1 and VEGFA blockade elicits antitumor response in hepatocellular carcinoma mediated by activated intratumoral CD8(+) exhausted-like T cells. Nat Commun. 2023;14(1):7709.
Bedard PL, et al. Small molecules, big impact: 20 years of targeted therapy in oncology. Lancet. 2020;395(10229):1078–88.
Tsimberidou AM, et al. Molecular tumour boards - current and future considerations for precision oncology. Nat Rev Clin Oncol. 2023;20(12):843–63.
Petroni G, et al. Targeting oncogene and non-oncogene addiction to inflame the tumour microenvironment. Nat Rev Drug Discov. 2022;21(6):440–62.
Sandhu S, et al. Phase 1b study of cobimetinib plus atezolizumab in patients with advanced BRAF(V600) wild-type melanoma progressing on prior anti-programmed death-1 therapy. Eur J Cancer. 2023;178:180–90.
Rozeman EA et al. IMPemBra: a phase 2 study comparing pembrolizumab with intermittent/short-term dual MAPK pathway inhibition plus pembrolizumab in patients with melanoma harboring the BRAFV600 mutation. J Immunother Cancer, 2023. 11(7).
Long GV, et al. Epacadostat plus Pembrolizumab versus placebo plus pembrolizumab in patients with unresectable or metastatic melanoma (ECHO-301/KEYNOTE-252): a phase 3, randomised, double-blind study. Lancet Oncol. 2019;20(8):1083–97.
Dummer R, et al. Randomized Phase III Trial evaluating Spartalizumab Plus Dabrafenib and Trametinib for BRAF V600-Mutant Unresectable or metastatic melanoma. J Clin Oncol. 2022;40(13):1428–38.
!!!. INVALID CITATION !!!.
Ascierto PA, et al. Overall survival with first-line atezolizumab in combination with vemurafenib and cobimetinib in BRAF(V600) mutation-positive advanced melanoma (IMspire150): second interim analysis of a multicentre, randomised, phase 3 study. Lancet Oncol. 2023;24(1):33–44.
Gutzmer R, et al. Atezolizumab, vemurafenib, and cobimetinib as first-line treatment for unresectable advanced BRAF(V600) mutation-positive melanoma (IMspire150): primary analysis of the randomised, double-blind, placebo-controlled, phase 3 trial. Lancet. 2020;395(10240):1835–44.
Dummer R, et al. Atezolizumab, vemurafenib, and cobimetinib in patients with melanoma with CNS metastases (TRICOTEL): a multicentre, open-label, single-arm, phase 2 study. Lancet Oncol. 2023;24(12):e461–71.
Ascierto PA, et al. Sequencing of Ipilimumab Plus Nivolumab and Encorafenib Plus Binimetinib for untreated BRAF-Mutated metastatic melanoma (SECOMBIT): a Randomized, Three-Arm, open-label phase II trial. J Clin Oncol. 2023;41(2):212–21.
Ascierto PA, et al. Sequential immunotherapy and targeted therapy for metastatic BRAF V600 mutated melanoma: 4-year survival and biomarkers evaluation from the phase II SECOMBIT trial. Nat Commun. 2024;15(1):146.
Atkins MB, et al. Combination Dabrafenib and Trametinib Versus Combination Nivolumab and Ipilimumab for patients with Advanced BRAF-Mutant Melanoma: the DREAMseq Trial-ECOG-ACRIN EA6134. J Clin Oncol. 2023;41(2):186–97.
Long GV, et al. Antitumor activity of ipilimumab or BRAF ± MEK inhibition after pembrolizumab treatment in patients with advanced melanoma: analysis from KEYNOTE-006. Ann Oncol. 2022;33(2):204–15.
Robert C, et al. Biomarkers of treatment benefit with atezolizumab plus vemurafenib plus cobimetinib in BRAF(V600) mutation-positive melanoma. Ann Oncol. 2022;33(5):544–55.
Dummer R, et al. Combined PD-1, BRAF and MEK inhibition in advanced BRAF-mutant melanoma: safety run-in and biomarker cohorts of COMBI-i. Nat Med. 2020;26(10):1557–63.
Reiss KA, et al. Niraparib plus Nivolumab or Niraparib plus Ipilimumab in patients with platinum-sensitive advanced pancreatic cancer: a randomised, phase 1b/2 trial. Lancet Oncol. 2022;23(8):1009–20.
He K, et al. MRTX-500 phase 2 trial: Sitravatinib with Nivolumab in patients with nonsquamous NSCLC progressing on or after checkpoint inhibitor therapy or chemotherapy. J Thorac Oncol. 2023;18(7):907–21.
Borghaei H, et al. SAPPHIRE: phase III study of sitravatinib plus nivolumab versus docetaxel in advanced nonsquamous non-small-cell lung cancer. Ann Oncol. 2024;35(1):66–76.
Masuda J et al. Efficacy, safety, and biomarker analysis of nivolumab in combination with abemaciclib plus endocrine therapy in patients with HR-positive HER2-negative metastatic breast cancer: a phase II study (WJOG11418B NEWFLAME trial). J Immunother Cancer, 2023. 11(9).
Xu Q, et al. Efficacy and safety of Sintilimab Plus Anlotinib for PD-L1-Positive recurrent or metastatic cervical Cancer: a Multicenter, Single-Arm, prospective phase II trial. J Clin Oncol. 2022;40(16):1795–805.
Donato C, et al. Hypoxia triggers the intravasation of clustered circulating Tumor cells. Cell Rep. 2020;32(10):108105.
Zhang L, et al. Endothelial DGKG promotes tumor angiogenesis and immune evasion in hepatocellular carcinoma. J Hepatol. 2024;80(1):82–98.
Cao J, et al. Decylubiquinone suppresses breast cancer growth and metastasis by inhibiting angiogenesis via the ROS/p53/ BAI1 signaling pathway. Angiogenesis. 2020;23(3):325–38.
Di Tacchio M, et al. Tumor Vessel normalization, Immunostimulatory Reprogramming, and Improved Survival in Glioblastoma with Combined Inhibition of PD-1, Angiopoietin-2, and VEGF. Cancer Immunol Res. 2019;7(12):1910–27.
Huinen ZR, et al. Anti-angiogenic agents - overcoming tumour endothelial cell anergy and improving immunotherapy outcomes. Nat Rev Clin Oncol. 2021;18(8):527–40.
Zemek RM, et al. Exploiting temporal aspects of cancer immunotherapy. Nat Rev Cancer. 2024;24(7):480–97.
Rini BI, et al. Pembrolizumab plus Axitinib versus Sunitinib for Advanced Renal-Cell Carcinoma. N Engl J Med. 2019;380(12):1116–27.
Joshi UM et al. A phase II trial of nivolumab plus axitinib in patients with anti-PD1 refractory advanced melanoma. 2024. 42(16_suppl): p. TPS9600–9600.
Finn RS, et al. Atezolizumab plus Bevacizumab in Unresectable Hepatocellular Carcinoma. N Engl J Med. 2020;382(20):1894–905.
Qin S, et al. Camrelizumab plus Rivoceranib versus Sorafenib as first-line therapy for unresectable hepatocellular carcinoma (CARES-310): a randomised, open-label, international phase 3 study. Lancet. 2023;402(10408):1133–46.
Saba NF, et al. Pembrolizumab and cabozantinib in recurrent metastatic head and neck squamous cell carcinoma: a phase 2 trial. Nat Med. 2023;29(4):880–7.
Cheng H, et al. Camrelizumab plus Apatinib in patients with high-risk chemorefractory or relapsed gestational trophoblastic neoplasia (CAP 01): a single-arm, open-label, phase 2 trial. Lancet Oncol. 2021;22(11):1609–17.
Pan QZ, et al. XELOX (capecitabine plus oxaliplatin) plus bevacizumab (anti-VEGF-A antibody) with or without adoptive cell immunotherapy in the treatment of patients with previously untreated metastatic colorectal cancer: a multicenter, open-label, randomized, controlled, phase 3 trial. Signal Transduct Target Ther. 2024;9(1):79.
Yuan L, et al. Camrelizumab combined with apatinib in patients with first-line platinum-resistant or PD-1 inhibitor resistant recurrent/metastatic nasopharyngeal carcinoma: a single-arm, phase 2 trial. Nat Commun. 2023;14(1):4893.
Motzer RJ, et al. Nivolumab plus Cabozantinib versus sunitinib in first-line treatment for advanced renal cell carcinoma (CheckMate 9ER): long-term follow-up results from an open-label, randomised, phase 3 trial. Lancet Oncol. 2022;23(7):888–98.
Wang D, et al. Intraparticle double-scattering-decoded sonogenetics for augmenting Immune Checkpoint Blockade and CAR-T therapy. Adv Sci (Weinh). 2022;9(32):e2203106.
Zhou Z, et al. Deciphering the tumor immune microenvironment from a multidimensional omics perspective: insight into next-generation CAR-T cell immunotherapy and beyond. Mol Cancer. 2024;23(1):131.
Chen N, et al. CAR T-cell intrinsic PD-1 checkpoint blockade: a two-in-one approach for solid tumor immunotherapy. Oncoimmunology. 2017;6(2):e1273302.
Cheng K et al. 4-1BB-Based CAR T cells effectively reverse exhaustion and enhance the Anti-tumor Immune Response through Autocrine PD-L1 scFv antibody. Int J Mol Sci, 2023. 24(4).
Hu W, et al. CRISPR/Cas9-mediated PD-1 disruption enhances human mesothelin-targeted CAR T cell effector functions. Cancer Immunol Immunother. 2019;68(3):365–77.
Cherkassky L, et al. Human CAR T cells with cell-intrinsic PD-1 checkpoint blockade resist tumor-mediated inhibition. J Clin Invest. 2016;126(8):3130–44.
Rafiq S, et al. Targeted delivery of a PD-1-blocking scFv by CAR-T cells enhances anti-tumor efficacy in vivo. Nat Biotechnol. 2018;36(9):847–56.
Gulati P, et al. Aberrant lck Signal via CD28 Costimulation augments Antigen-Specific functionality and Tumor Control by Redirected T Cells with PD-1 blockade in Humanized mice. Clin Cancer Res. 2018;24(16):3981–93.
Yamaguchi Y et al. PD-L1 blockade restores CAR T cell activity through IFN-γ-regulation of CD163 + M2 macrophages. J Immunother Cancer, 2022. 10(6).
Gargett T, et al. GD2-specific CAR T cells undergo potent activation and deletion following Antigen Encounter but can be protected from activation-induced cell death by PD-1 blockade. Mol Ther. 2016;24(6):1135–49.
Lee YH, et al. PD-1 and TIGIT downregulation distinctly affect the effector and early memory phenotypes of CD19-targeting CAR T cells. Mol Ther. 2022;30(2):579–92.
Ouyang W et al. PD-1 downregulation enhances CAR-T cell antitumor efficiency by preserving a cell memory phenotype and reducing exhaustion. J Immunother Cancer, 2024. 12(4).
Sengupta S, et al. Glycogen synthase kinase 3 inhibition lowers PD-1 expression, promotes long-term survival and memory generation in antigen-specific CAR-T cells. Cancer Lett. 2018;433:131–9.
Choi BD, et al. CRISPR-Cas9 disruption of PD-1 enhances activity of universal EGFRvIII CAR T cells in a preclinical model of human glioblastoma. J Immunother Cancer. 2019;7(1):304.
Chen J, et al. Target delivery of a PD-1-TREM2 scFv by CAR-T cells enhances anti-tumor efficacy in colorectal cancer. Mol Cancer. 2023;22(1):131.
Gray KD, et al. PD1 blockade enhances ICAM1-Directed CAR T therapeutic efficacy in advanced thyroid Cancer. Clin Cancer Res. 2020;26(22):6003–16.
Harrasser M, et al. Inducible localized delivery of an anti-PD-1 scFv enhances anti-tumor activity of ROR1 CAR-T cells in TNBC. Breast Cancer Res. 2022;24(1):39.
Hu Y, et al. Safety and efficacy of CRISPR-based non-viral PD1 locus specifically integrated anti-CD19 CAR-T cells in patients with relapsed or refractory Non-hodgkin’s lymphoma: a first-in-human phase I study. EClinicalMedicine. 2023;60:102010.
Heczey A, et al. CAR T cells administered in combination with lymphodepletion and PD-1 inhibition to patients with Neuroblastoma. Mol Ther. 2017;25(9):2214–24.
Liu H, et al. CD19-specific CAR T cells that Express a PD-1/CD28 chimeric switch-receptor are effective in patients with PD-L1-positive B-Cell lymphoma. Clin Cancer Res. 2021;27(2):473–84.
Adusumilli PS, et al. A phase I Trial of Regional Mesothelin-targeted CAR T-cell therapy in patients with malignant Pleural Disease, in combination with the Anti-PD-1 Agent Pembrolizumab. Cancer Discov. 2021;11(11):2748–63.
Wang Y, et al. Anti-PD-1 antibody armored γδ T cells enhance anti-tumor efficacy in ovarian cancer. Signal Transduct Target Ther. 2023;8(1):399.
Huang SW, et al. BiTE-Secreting CAR-γδT as a dual targeting strategy for the treatment of solid tumors. Adv Sci (Weinh). 2023;10(17):e2206856.
Chang Y, et al. Engineered human pluripotent stem cell-derived natural killer cells with PD-L1 responsive immunological memory for enhanced immunotherapeutic efficacy. Bioact Mater. 2023;27:168–80.
Wang F, et al. Combined treatment with anti-PSMA CAR NK-92 cell and anti-PD-L1 monoclonal antibody enhances the antitumour efficacy against castration-resistant prostate cancer. Clin Transl Med. 2022;12(6):e901.
Liu WN, et al. Successful targeting of PD-1/PD-L1 with chimeric antigen receptor-natural killer cells and nivolumab in a humanized mouse cancer model. Sci Adv. 2022;8(47):eadd1187.
Creelan BC, et al. Tumor-infiltrating lymphocyte treatment for anti-PD-1-resistant metastatic lung cancer: a phase 1 trial. Nat Med. 2021;27(8):1410–8.
Melcher A, Harrington K, Vile R. Oncolytic virotherapy as immunotherapy. Science. 2021;374(6573):1325–6.
Greig SL, Laherparepvec T. First Global Approval Drugs. 2016;76(1):147–54.
Poh A. First oncolytic viral therapy for Melanoma. Cancer Discov. 2016;6(1):6.
Chesney J, et al. Randomized, open-label phase II study evaluating the efficacy and safety of Talimogene Laherparepvec in Combination with Ipilimumab Versus Ipilimumab alone in patients with Advanced, Unresectable Melanoma. J Clin Oncol. 2018;36(17):1658–67.
Chesney JA et al. Talimogene Laherparepvec in combination with ipilimumab versus ipilimumab alone for advanced melanoma: 5-year final analysis of a multicenter, randomized, open-label, phase II trial. J Immunother Cancer, 2023. 11(5).
Chesney JA, et al. Randomized, Double-Blind, Placebo-Controlled, global phase III trial of Talimogene Laherparepvec Combined with Pembrolizumab for Advanced Melanoma. J Clin Oncol. 2023;41(3):528–40.
Middleton M, et al. 422 an open-label, multicenter, phase 1/2 clinical trial of RP1, an enhanced potency oncolytic HSV, combined with nivolumab: updated results from the skin cancer cohorts. J Immunother Cancer. 2020;8:A448–448.
Chmielowski B et al. Initial efficacy and safety of RP1 + nivolumab in patients with anti–PD-1–failed melanoma from the ongoing phase 1/2 IGNYTE study. 2023. 41(16_suppl): pp. 9509–9509.
Lenz H-J et al. An open-label clinical trial of RP2 and RP3 oncolytic immunotherapy in combination with atezolizumab plus bevacizumab for the treatment of patients with advanced colorectal carcinoma. 2024. 42(3_suppl): p. TPS228–228.
Gujar S et al. Tutorial: design, production and testing of oncolytic viruses for cancer immunotherapy. Nat Protoc, 2024.
Phase 1 Study of Intratumoral Administration of JNJ-87704916, an Oncolytic Virus, as Monotherapy and in Combination for Advanced Solid Tumors. 2024.
Observation of the Efficacy and Safety of Oncolytic Adenovirus Injection Combined With Programmed Death Receptor Inhibitors in Treatment of Advanced Malignant Pleural Mesothelioma: a Single Center, Prospective, Case Registration Study. 2023.
neoBREASTIM:. A phase 2 study of Atezolizumab Plus RP1 Oncolytic Immunotherapy in the NeoAdjuvant setting of Triple-negative breast Cancer (TNBC). Editors: and A.G. Roche Pharma; 2023. I. Replimune.
Two- A. part, Phase I/Ib, Open-Label, Dose-escalation Trial of Tumor Necrosis Factor Alpha and Interleukin-2 Coding Oncolytic Adenovirus (TILT-123) in Combination With Pembrolizumab (Phase I Part) and Pembrolizumab and Pegylated Liposomal Doxorubicin (Phase Ib Part) in Patients With Platinum Resistant or Refractory Ovarian Cancer, S. Merck and L.L.C. Dohme, Editors. 2022.
Phase IIS. Center Open-Label Single-Arm Study of the Safety and Efficacy of Oncolytic Adenovirus H101 Combined With PD-1 Inhibitor in Patients With Non-muscle-invasive Bladder Cancer Who Failed BCG Therapy. 2022.
Dohme, editor. IIa study of intra-tumoral BT-001 (TG6030) administered alone and in Combination with Pembrolizumab in patients with cutaneous or, Subcutaneous lesions or easily Injectable Lymph nodes of Metastatic/Advanced solid tumors. S. Merck, and L.L.C.: A.B. BioInvent International; 2021. A Phase I/.
Phase I. Study of MEM-288 Oncolytic Virus Alone and in Combination With Nivolumab in Solid Tumors Including Non-Small Cell Lung Cancer (NSCLC), I. Duke Cancer, H.L.M.C. Center, and I. Research, Editors. 2021.
An Open-Label, Multicenter, Phase 1/2 Study of RP1 as a Single Agent and in Combination With PD1 Blockade in Patients With Solid Tumors. 2018.
Terai M, Sato T. Individualised neoantigen cancer vaccine therapy. Lancet. 2024;403(10427):590–1.
Saxena M, et al. Therapeutic cancer vaccines. Nat Rev Cancer. 2021;21(6):360–78.
Fucikova J, et al. An autologous dendritic cell vaccine promotes anticancer immunity in patients with ovarian Cancer with low mutational Burden and Cold tumors. Clin Cancer Res. 2022;28(14):3053–65.
Rojas LA, et al. Personalized RNA neoantigen vaccines stimulate T cells in pancreatic cancer. Nature. 2023;618(7963):144–50.
Sahin U, et al. An RNA vaccine drives immunity in checkpoint-inhibitor-treated melanoma. Nature. 2020;585(7823):107–12.
Rappaport AR, et al. A shared neoantigen vaccine combined with immune checkpoint blockade for advanced metastatic solid tumors: phase 1 trial interim results. Nat Med. 2024;30(4):1013–22.
Zhou Y, et al. A pilot study of multi-antigen stimulated cell therapy-I plus camrelizumab and apatinib in patients with advanced bone and soft-tissue sarcomas. BMC Med. 2023;21(1):470.
Ellingsen EB, et al. Clinical activity of combined telomerase vaccination and Pembrolizumab in Advanced Melanoma: results from a phase I Trial. Clin Cancer Res. 2023;29(16):3026–36.
McNeel DG et al. Phase 2 trial of a DNA vaccine (pTVG-HP) and nivolumab in patients with castration-sensitive non-metastatic (M0) prostate cancer. J Immunother Cancer, 2023. 11(12).
Autio KA et al. First-in-human, phase 1 study of PF-06753512, a vaccine-based immunotherapy regimen (VBIR), in non-metastatic hormone-sensitive biochemical recurrence and metastatic castration-resistant prostate cancer (mCRPC). J Immunother Cancer, 2023. 11(3).
Massarelli E, et al. Combining Immune Checkpoint Blockade and Tumor-specific vaccine for patients with Incurable Human Papillomavirus 16-Related Cancer: a phase 2 clinical trial. JAMA Oncol. 2019;5(1):67–73.
Youn JW, et al. Pembrolizumab plus GX-188E therapeutic DNA vaccine in patients with HPV-16-positive or HPV-18-positive advanced cervical cancer: interim results of a single-arm, phase 2 trial. Lancet Oncol. 2020;21(12):1653–60.
Stein-Thoeringer CK, et al. A non-antibiotic-disrupted gut microbiome is associated with clinical responses to CD19-CAR-T cell cancer immunotherapy. Nat Med. 2023;29(4):906–16.
Huang J, et al. Ginseng polysaccharides alter the gut microbiota and kynurenine/tryptophan ratio, potentiating the antitumour effect of antiprogrammed cell death 1/programmed cell death ligand 1 (anti-PD-1/PD-L1) immunotherapy. Gut. 2022;71(4):734–45.
Matson V, et al. The commensal microbiome is associated with anti-PD-1 efficacy in metastatic melanoma patients. Science. 2018;359(6371):104–8.
Routy B, et al. Gut microbiome influences efficacy of PD-1-based immunotherapy against epithelial tumors. Science. 2018;359(6371):91–7.
McCulloch JA, et al. Intestinal microbiota signatures of clinical response and immune-related adverse events in melanoma patients treated with anti-PD-1. Nat Med. 2022;28(3):545–56.
Baruch EN, et al. Fecal microbiota transplant promotes response in immunotherapy-refractory melanoma patients. Science. 2021;371(6529):602–9.
Glitza IC, et al. Randomized Placebo-Controlled, biomarker-stratified phase ib Microbiome Modulation in Melanoma: impact of Antibiotic Preconditioning on Microbiome and Immunity. Cancer Discov. 2024;14(7):1161–75.
Wu H, et al. Dynamic microbiome and metabolome analyses reveal the interaction between gut microbiota and anti-PD-1 based immunotherapy in hepatocellular carcinoma. Int J Cancer. 2022;151(8):1321–34.
Alspach E, et al. MHC-II neoantigens shape tumour immunity and response to immunotherapy. Nature. 2019;574(7780):696–701.
Dersh D, et al. Genome-wide screens identify lineage- and tumor-specific genes modulating MHC-I- and MHC-II-Restricted immunosurveillance of human lymphomas. Immunity. 2021;54(1):116–e13110.
Duong E, et al. Type I interferon activates MHC class I-dressed CD11b(+) conventional dendritic cells to promote protective anti-tumor CD8(+) T cell immunity. Immunity. 2022;55(2):308–e3239.
Ardeniz Ö, et al. β2-Microglobulin deficiency causes a complex immunodeficiency of the innate and adaptive immune system. J Allergy Clin Immunol. 2015;136(2):392–401.
Burr ML, et al. An evolutionarily conserved function of polycomb silences the MHC Class I Antigen Presentation Pathway and enables Immune Evasion in Cancer. Cancer Cell. 2019;36(4):385–e4018.
Gu SS, et al. Therapeutically increasing MHC-I expression potentiates Immune Checkpoint Blockade. Cancer Discov. 2021;11(6):1524–41.
Du W, et al. Loss of Optineurin Drives Cancer Immune Evasion via Palmitoylation-Dependent IFNGR1 lysosomal sorting and degradation. Cancer Discov. 2021;11(7):1826–43.
Yamamoto K, et al. Autophagy promotes immune evasion of pancreatic cancer by degrading MHC-I. Nature. 2020;581(7806):100–5.
Bolivar AM, et al. Genomic Landscape of Lynch Syndrome Colorectal Neoplasia identifies Shared Mutated Neoantigens for Immunoprevention. Gastroenterology. 2024;166(5):787–e80111.
McGrail DJ, et al. Proteome instability is a therapeutic vulnerability in Mismatch Repair-Deficient Cancer. Cancer Cell. 2020;37(3):371–e38612.
Marabelle A, 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(1):1–10.
Diaz LA Jr., et al. Pembrolizumab versus chemotherapy for microsatellite instability-high or mismatch repair-deficient metastatic colorectal cancer (KEYNOTE-177): final analysis of a randomised, open-label, phase 3 study. Lancet Oncol. 2022;23(5):659–70.
Hogg SJ, et al. Targeting the epigenetic regulation of antitumour immunity. Nat Rev Drug Discov. 2020;19(11):776–800.
Mittal P, Roberts CWM. The SWI/SNF complex in cancer - biology, biomarkers and therapy. Nat Rev Clin Oncol. 2020;17(7):435–48.
Chiappinelli KB, et al. Inhibiting DNA methylation causes an Interferon Response in Cancer via dsRNA. Including Endogenous Retroviruses Cell. 2015;162(5):974–86.
Cañadas I, et al. Tumor innate immunity primed by specific interferon-stimulated endogenous retroviruses. Nat Med. 2018;24(8):1143–50.
Goyal A, et al. DNMT and HDAC inhibition induces immunogenic neoantigens from human endogenous retroviral element-derived transcripts. Nat Commun. 2023;14(1):6731.
Ng KW, et al. Antibodies against endogenous retroviruses promote lung cancer immunotherapy. Nature. 2023;616(7957):563–73.
Weinberg DN, et al. The histone mark H3K36me2 recruits DNMT3A and shapes the intergenic DNA methylation landscape. Nature. 2019;573(7773):281–6.
Zhang B, et al. M(6)a regulator-mediated methylation modification patterns and tumor microenvironment infiltration characterization in gastric cancer. Mol Cancer. 2020;19(1):53.
Martins FC, et al. Clonal somatic copy number altered driver events inform drug sensitivity in high-grade serous ovarian cancer. Nat Commun. 2022;13(1):6360.
Fang H, et al. Combination of epigenetic regulation with gene therapy-mediated immune checkpoint blockade induces anti-tumour effects and immune response in vivo. Nat Commun. 2021;12(1):6742.
Lai J, et al. Zebularine elevates STING expression and enhances cGAMP cancer immunotherapy in mice. Mol Ther. 2021;29(5):1758–71.
Pan Y, et al. EHBP1L1 drives Immune Evasion in Renal Cell Carcinoma through binding and stabilizing JAK1. Adv Sci (Weinh). 2023;10(11):e2206792.
Tong Z et al. Activation of the cGAS-STING-IRF3 Axis by Type I and II interferons contributes to host defense. Adv Sci (Weinh), 2024: p. e2308890.
Wang S, et al. Down-regulation of DNA key protein-FEN1 inhibits OSCC growth by affecting immunosuppressive phenotypes via IFN-γ/JAK/STAT-1. Int J Oral Sci. 2023;15(1):17.
Parsons MJ, Tammela T, Dow LE. WNT as a driver and dependency in Cancer. Cancer Discov. 2021;11(10):2413–29.
Han T, et al. Lineage reversion drives WNT independence in Intestinal Cancer. Cancer Discov. 2020;10(10):1590–609.
Xie G, et al. Decreased expression of ATF3, orchestrated by β-catenin/TCF3, mir-17-5p and HOXA11-AS, promoted gastric cancer progression via increased β-catenin and CEMIP. Exp Mol Med. 2021;53(11):1706–22.
Taniue K, et al. ASBEL-TCF3 complex is required for the tumorigenicity of colorectal cancer cells. Proc Natl Acad Sci U S A. 2016;113(45):12739–44.
Katoh Y et al. Inhibition of stearoyl-CoA desaturase 1 (SCD1) enhances the antitumor T cell response through regulating β-catenin signaling in cancer cells and ER stress in T cells and synergizes with anti-PD-1 antibody. J Immunother Cancer, 2022. 10(7).
Zhou L, et al. Hypoxia-induced lncRNA STEAP3-AS1 activates Wnt/β-catenin signaling to promote colorectal cancer progression by preventing m(6)A-mediated degradation of STEAP3 mRNA. Mol Cancer. 2022;21(1):168.
Jin C, et al. Acetyltransferase NAT10 regulates the Wnt/β-catenin signaling pathway to promote colorectal cancer progression via Ac(4)C acetylation of KIF23 mRNA. J Exp Clin Cancer Res. 2022;41(1):345.
Huang Q, et al. WNT7A promotes tumorigenesis of head and neck squamous cell carcinoma via activating FZD7/JAK1/STAT3 signaling. Int J Oral Sci. 2024;16(1):7.
Exposito F, et al. PTEN loss confers resistance to Anti-PD-1 therapy in Non-small Cell Lung Cancer by Increasing Tumor Infiltration of Regulatory T Cells. Cancer Res. 2023;83(15):2513–26.
Yu X, et al. Omics analyses of a somatic Trp53(R245W/+) breast cancer model identify cooperating driver events activating PI3K/AKT/mTOR signaling. Proc Natl Acad Sci U S A. 2022;119(45):e2210618119.
Duchatel RJ et al. PI3K/mTOR is a therapeutically targetable genetic dependency in diffuse intrinsic pontine glioma. J Clin Invest, 2024. 134(6).
Lin YX et al. Reactivation of the tumor suppressor PTEN by mRNA nanoparticles enhances antitumor immunity in preclinical models. Sci Transl Med, 2021. 13(599).
Yuan S, et al. Ras drives malignancy through stem cell crosstalk with the microenvironment. Nature. 2022;612(7940):555–63.
Johnson C, Burkhart DL, Haigis KM. Classification of KRAS-Activating mutations and the implications for therapeutic intervention. Cancer Discov. 2022;12(4):913–23.
Jänne PA, et al. Adagrasib in Non-small-cell Lung Cancer harboring a KRAS(G12C) mutation. N Engl J Med. 2022;387(2):120–31.
Bekaii-Saab TS, et al. Adagrasib in Advanced Solid tumors harboring a KRAS(G12C) mutation. J Clin Oncol. 2023;41(25):4097–106.
Soula M et al. Glycosphingolipid synthesis mediates immune evasion in KRAS-driven cancer. Nature, 2024.
Puyalto A, et al. Trametinib sensitizes KRAS-mutant lung adenocarcinoma tumors to PD-1/PD-L1 axis blockade via Id1 downregulation. Mol Cancer. 2024;23(1):78.
Zhou Q et al. Efficacy and safety of KRAS G12C inhibitor IBI351 monotherapy in patients with advanced non-small cell lung cancer: results from a phase 2 pivotal study. J Thorac Oncol, 2024.
Cho SF, Anderson KC, Tai YT. Microenvironment is a key determinant of Immune checkpoint inhibitor response. Clin Cancer Res. 2022;28(8):1479–81.
Van Laethem JL, et al. Combining CD40 agonist mitazalimab with mFOLFIRINOX in previously untreated metastatic pancreatic ductal adenocarcinoma (OPTIMIZE-1): a single-arm, multicentre phase 1b/2 study. Lancet Oncol. 2024;25(7):853–64.
Barkley D, et al. Cancer cell states recur across tumor types and form specific interactions with the tumor microenvironment. Nat Genet. 2022;54(8):1192–201.
Dongre A, et al. Direct and indirect regulators of epithelial-mesenchymal transition-mediated immunosuppression in breast carcinomas. Cancer Discov. 2021;11(5):1286–305.
Plaschka M et al. ZEB1 transcription factor promotes immune escape in melanoma. J Immunother Cancer, 2022. 10(3).
Ma C, et al. Pan-cancer spatially resolved single-cell analysis reveals the crosstalk between cancer-associated fibroblasts and tumor microenvironment. Mol Cancer. 2023;22(1):170.
Gu Y, et al. Genome-wide CRISPR screens define determinants of epithelial-mesenchymal transition mediated immune evasion by pancreatic cancer cells. Sci Adv. 2023;9(28):eadf9915.
Long L, et al. Regulating lactate-related immunometabolism and EMT reversal for colorectal cancer liver metastases using shikonin targeted delivery. J Exp Clin Cancer Res. 2023;42(1):117.
Wu N, et al. LINC00941 promotes CRC metastasis through preventing SMAD4 protein degradation and activating the TGF-β/SMAD2/3 signaling pathway. Cell Death Differ. 2021;28(1):219–32.
Horn LA et al. Remodeling the tumor microenvironment via blockade of LAIR-1 and TGF-β signaling enables PD-L1-mediated tumor eradication. J Clin Invest, 2022. 132(8).
Honda CK, et al. Cancer-associated fibroblast spatial heterogeneity and EMILIN1 expression in the tumor microenvironment modulate TGF-β activity and CD8(+) T-cell infiltration in breast cancer. Theranostics. 2024;14(5):1873–85.
Goswami MT, et al. Regulation of complement-dependent cytotoxicity by TGF-β-induced epithelial-mesenchymal transition. Oncogene. 2016;35(15):1888–98.
Sun CC, et al. FOXC1-mediated LINC00301 facilitates tumor progression and triggers an immune-suppressing microenvironment in non-small cell lung cancer by regulating the HIF1α pathway. Genome Med. 2020;12(1):77.
Lasser SA, et al. Myeloid-derived suppressor cells in cancer and cancer therapy. Nat Rev Clin Oncol. 2024;21(2):147–64.
De Henau O, et al. Overcoming resistance to checkpoint blockade therapy by targeting PI3Kγ in myeloid cells. Nature. 2016;539(7629):443–7.
Xu H, et al. Flagella of Tumor-Targeting Bacteria Trigger Local Hemorrhage to Reprogram Tumor-Associated macrophages for Improved Antitumor Therapy. Adv Mater. 2023;35(38):e2303357.
Wang H, et al. The promising role of tumor-associated macrophages in the treatment of cancer. Drug Resist Updat. 2024;73:101041.
Cortez JT, et al. CRISPR screen in regulatory T cells reveals modulators of Foxp3. Nature. 2020;582(7812):416–20.
Zagorulya M, et al. Tissue-specific abundance of interferon-gamma drives regulatory T cells to restrain DC1-mediated priming of cytotoxic T cells against lung cancer. Immunity. 2023;56(2):386–e40510.
Moreno Ayala MA, et al. CXCR3 expression in regulatory T cells drives interactions with type I dendritic cells in tumors to restrict CD8(+) T cell antitumor immunity. Immunity. 2023;56(7):1613–e16305.
Prelaj A, et al. Artificial intelligence for predictive biomarker discovery in immuno-oncology: a systematic review. Ann Oncol. 2024;35(1):29–65.
Che G, et al. Circumventing drug resistance in gastric cancer: a spatial multi-omics exploration of chemo and immuno-therapeutic response dynamics. Drug Resist Updat. 2024;74:101080.
Bao X, et al. A multiomics analysis-assisted deep learning model identifies a macrophage-oriented module as a potential therapeutic target in colorectal cancer. Cell Rep Med. 2024;5(2):101399.
Funding
This work was supported by the 1.3.5 Project for Disciplines of Excellence, West China Hospital, Sichuan University (Grant No. ZYJC21043), and Social Development Science and Technology Project of Sichuan Province on Science and Technology (2023YFS0111).
Author information
Authors and Affiliations
Contributions
ML contributed to conception and design of the manuscript. JW participated in the literature search, drafted the manuscript, and designed the figures. WKL, PFZ, FKG and ML supervised and edited the manuscript. Final draft read and approved by all authors.
Corresponding author
Ethics declarations
Ethics approval and consent to participate
Not applicable.
Consent for publication
Not applicable.
Competing interests
The authors declare no competing interests.
Additional information
Publisher’s note
Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.
Rights and permissions
Open Access This article is licensed under a Creative Commons Attribution-NonCommercial-NoDerivatives 4.0 International License, which permits any non-commercial use, sharing, distribution and reproduction in any medium or format, as long as you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons licence, and indicate if you modified the licensed material. You do not have permission under this licence to share adapted material derived from this article or parts of it. The images or other third party material in this article are included in the article’s Creative Commons licence, unless indicated otherwise in a credit line to the material. If material is not included in the article’s Creative Commons licence and your intended use is not permitted by statutory regulation or exceeds the permitted use, you will need to obtain permission directly from the copyright holder. To view a copy of this licence, visit http://creativecommons.org/licenses/by-nc-nd/4.0/.
About this article
Cite this article
Wei, J., Li, W., Zhang, P. et al. Current trends in sensitizing immune checkpoint inhibitors for cancer treatment. Mol Cancer 23, 279 (2024). https://doiorg.publicaciones.saludcastillayleon.es/10.1186/s12943-024-02179-5
Received:
Accepted:
Published:
DOI: https://doiorg.publicaciones.saludcastillayleon.es/10.1186/s12943-024-02179-5