Open Access

Beyond the tumor microenvironment: Orchestrating systemic T‑cell response for next‑generation cancer immunotherapy (Review)

  • Authors:
    • Xiaohong Lyu
    • Jiashu Han
    • Chen Lin
    • Yidong Zhou
    • Weibin Wang
  • View Affiliations

  • Published online on: June 13, 2025     https://doi.org/10.3892/ijo.2025.5762
  • Article Number: 56
  • Copyright: © Lyu et al. This is an open access article distributed under the terms of Creative Commons Attribution License.

Metrics: Total Views: 0 (Spandidos Publications: | PMC Statistics: )
Total PDF Downloads: 0 (Spandidos Publications: | PMC Statistics: )


Abstract

Immune checkpoint blockade therapy has revolutionized cancer treatment, yet its clinical efficacy remains limited to a subset of patients with specific tumor types. The present review provides a comprehensive analysis of T cell‑mediated antitumor immunity from both local and systemic perspectives, with particular emphasis on CD8+ T cells as primary effectors. The review discusses how the complex trafficking between the tumor microenvironment (TME), surrounding lymphoid tissues and peripheral circulation creates multiple opportunities for tumors to evade immune surveillance. Within the TME, T‑cell exclusion mechanisms, antigen specificity and the spectrum of T‑cell exhaustion states, from progenitor exhausted T cells to terminally exhausted T‑cell phenotypes, are reviewed. Beyond the local TME, the crucial roles of tumor‑draining lymph nodes and tertiary lymphoid structures in maintaining sustainable antitumor immunity, as well as the significance of circulating T cells as both biomarkers and therapeutic targets, are analyzed. This systemic perspective provides insights into the dynamic nature of antitumor immunity and suggests potential strategies for next‑generation immunotherapies, including combination approaches targeting multiple immune compartments to achieve optimal therapeutic outcomes.

Introduction

Immune checkpoint blockade (ICB) therapy has demonstrated marked clinical efficacy, albeit limited to a subset of patients with specific solid tumor types, such as melanoma, non-small cell lung cancer (NSCLC), renal cell carcinoma and urothelial carcinoma. Extensive research has been conducted to elucidate the characteristics of responding patients and the underlying mechanisms, aiming to identify suitable candidates for precision therapy and potential therapeutic targets to overcome resistance (1). A previous study demonstrated that ICB therapy efficacy is associated with an immunologically 'hot' phenotype of tumors (2). However, individual parameters, including programmed death ligand 1 (PD-L1) expression, tumor mutational burden (TMB) and T-cell infiltration (3), have proven insufficient as predictive biomarkers for ICB response. Furthermore, given that the immune response is an inherently systemic process, localized ICB administration often fails to achieve complete tumor elimination through immune-mediated mechanisms, such as cytotoxic T-cell activation and infiltration, which are critical for targeting and destroying tumor cells (4,5).

As with all immune responses, antitumor T cells undergo a sequential process of priming, activation, circulation, recruitment, infiltration, effector function, and ultimately, resolution or memory formation. The intricate trafficking network among the local tumor microenvironment (TME), surrounding lymphoid tissues and peripheral circulation provides multiple opportunities for tumors to abrogate effective antitumor immunity, necessitating comprehensive strategies to address all immune escape mechanisms for successful antitumor immune responses (6).

The recent advancement in multi-omics analysis has provided deeper insights into the complexities of antitumor immunity. Therefore, focusing on CD8+ T cells as the principal effectors of antitumor immune responses, the present review examines the comprehensive tumor ecosystem beyond the local TME, with particular emphasis on the dynamic and systemic nature of antitumor immunity, to provide direction for future basic and translational research.

T cells in the TME

The TME is a complex ecosystem consisting of immune and stromal cells, along with bioactive molecules that influence tumor progression and immune responses (Fig. 1) (7). Key components include T cells, tumor cells, cancer-associated fibroblasts (CAFs), myeloid-derived suppressor cells and tumor endothelial cells (TECs). Additionally, the extracellular matrix (ECM) and stromal molecules, such as collagen, chemokines, cytokines and signaling molecules, significantly impact tumor growth and T-cell function (Fig. 2). Among these, CD8+ tumor-infiltrating lymphocytes (TILs) are established prognostic biomarkers and form the foundation of most immunotherapies (8). However, their therapeutic efficacy is limited by their inherent heterogeneity in composition, phenotype and function, which complicates their correlation with treatment outcomes.

Exclusion

Although high TIL levels generally predict better therapeutic outcomes (9), immune-excluded tumors exhibit poor responses due to T cells being trapped in the tumor stroma, unable to penetrate the tumor core. This phenomenon, often mediated by CAFs and TECs, results in limited T cell-tumor contact and suboptimal ICB efficacy (10-12). CAFs, originating from diverse sources such as mesenchymal cells, epithelial-mesenchymal transition and stromal activation, exhibit functional plasticity (13). CAFs polarize into inflammatory CAFs, which secrete immunosuppressive cytokines, or myofibroblastic CAFs, which create physical barriers through ECM remodeling (14). A small subset of CD74+ CAFs can also present antigens, directly modulating immune responses (15).

Similarly, TECs impede T-cell infiltration through abnormal vascular responses, including reduced adhesion molecule expression. Abnormal responses of blood vessel TECs to inflammatory cytokines and decreased expression of adhesion molecules impede T-cell recruitment and infiltration (16,17). Studies have demonstrated that high inflammation coupled with low vasculature correlates with optimal ICB responses, as confirmed by two independent investigations (18,19). Lymphatic TECs further exacerbate immune exclusion by attracting C-X-C chemokine receptor type 4 (CXCR4)+ T cells away from tumors via C-X-C motif chemokine ligand 12 (CXCL12) signaling (20). Collectively, these stromal cells establish chronic inflammatory niches that hinder effective immune responses. Recently, research (21) in hepatocellular carcinoma has revealed that pericancerous macrophages cross-present antigens to CD103+ CTLs via the endoplasmic reticulum-associated degradation machinery-mediated cytosolic pathway, subsequently activating the NLRP3 inflammasome in macrophages, thereby promoting the progression of hepatoma and immunotherapy resistance.

Specificity

T-cell specificity, driven by T-cell receptor (TCR) recognition of major histocompatibility complex-I:antigen complexes, underpins T-cell activation and antitumor immunity. TMB and neoantigen availability strongly correlate with ICB efficacy in immunologically 'hot' tumors such as melanoma and lung cancer (22-24). Conversely, in 'cold' tumors with low TMB, treatments such as chemotherapy, radiotherapy and tumor vaccines can enhance antigenicity, converting cold tumors into hot ones (25-28).

Despite these advances, most T cells in the TME are bystander T cells (Tbys) that lack tumor antigen recognition but still influence immune responses (29). Studies suggest that expressing Tby-specific antigens in tumor cells could harness their antitumor potential (30-32). By contrast, antigen-specific T cells (Tas), which recognize tumor neoantigens, are the primary effectors in immunotherapy (33). These cells often exhibit exhaustion due to chronic stimulation (34), characterized by markers such as CD39 (31,35,36,37), CD103 (38-40) and programmed cell death protein 1 (PD-1) (41,42). Emerging markers such as CXCL13 (43-47) and CD137 (48,49) further refine Tas identification and therapeutic targeting. Notably, the predominant expression of exhaustion markers among Tas markers emphasizes the critical need to address T-cell exhaustion in therapeutic strategies to maintain clonal expansion and effector function (50).

Exhaustion

Chronic antigen exposure in tumors induces T-cell exhaustion, marked by impaired proliferation, reduced cytokine production and elevated inhibitory receptor expression (51,52). T-cell exhaustion represents a complex process orchestrated by dynamic alterations and dysregulation in signaling pathways, transcription factors [including T cell factor 1 (TCF1), T-box expressed in T cells and thymocyte selection-associated high mobility group box protein (TOX)], epigenetic programs and metabolic adaptations (53,54). Exhausted T cells (Tex) comprise a heterogeneous population exhibiting a spectrum of phenotypic and functional characteristics (55). This spectrum is anchored by two distinct states: Progenitor Tex (Tex-prog), which maintain stem-like and proliferative properties, and terminally differentiated Tex (Tex-term).

Tex-prog

Tex-prog cells are characterized by high expression of TCF1 (56,57), a transcription factor essential for maintaining stemness and proliferative capacity. These cells respond well to ICB (58-62) and IL-2 (63-65) therapies. Tex-prog cells are considered primary mediators in chronic infection and ICB response (66-69), serving as primary mediators of long-term immune responses (70). The presence of the cells correlates with improved survival in melanoma and other cancer type. Notably, CXCL13+ T cells also exhibit Tex-prog characteristics (71).

Tex-term

Tex-term, defined by TOX activity (72-75) and multiple inhibitory checkpoints [e.g., PD-1 and lymphocyte activation gene 3 (LAG3)] (76-78), exhibit irreversible epigenetic changes, rendering them resistant to ICB (69). Under hypoxic conditions, Tex-term suppress immune responses via CD39-adenosine signaling (79), contributing to immunotherapy resistance. Strategies targeting Tex-term-associated pathways, such as P-selectin glycoprotein ligand-1 (80) and CD39 (81,82), are under investigation.

Intermediate Tex (Tex-int cells)

Tex-int cells occupy an intermediate state between functional effector T cells and Tex-term. These cells retain proliferative capacity and respond robustly to ICB, serving as a 'stem-like' reservoir for sustained immune responses (68,69). The abundance of these cells correlates with improved clinical outcomes, making them promising targets for combination therapies such as vaccines and chimeric antigen receptor (CAR)-T cells (53).

Key mechanisms regulating T-cell exhaustion

T-cell exhaustion results from persistent antigen stimulation and is regulated by a complex interplay of signaling pathways (71), transcription factors and epigenetic programs. Factors such as mechanistic target of rapamycin (mTOR), transforming growth factor-β (TGF-β) and PD-1 signaling influence transcriptional regulators, such as basic leucine zipper transcription factor ATF-like (BATF), TOX and TCF1, which maintain a delicate balance between T-cell activation and exhaustion (83,84). Briefly, upstream molecular pathways, such as mTOR and TGFβ (85), PD1 signaling (86) and IL-10 stimulation (87), orchestrate the activities of transcription factors myeloblastosis (88), activator protein 1 (61,89), BATF (90-92), BTB and CNC Homology 2 (BACH2) (90), special AT-rich sequence-binding protein 1 (93), interferon regulatory factor 4 and nuclear factor of activated T cells cytoplasmic 1 (91,92,94). Multiple aspects of a T cell are kept in a fine balance by these regulators; for example, methylcytosine dioxygenase TET2 guards against BATF3-induced CAR-T cell proliferation and ensuing genomic instability (95,96). With a more complete understanding of this epigenetic programming, it is possible to fine tune T-cell immunity and exhaustion while keeping proliferative responses and general inflammation in check.

Systemic T-cell response

T cells in lymph nodes

The systemic T-cell response to ICB is driven by two key mechanisms: The expansion of pre-existing T-cell clones and the infiltration of new clonotypes, a process termed clonal rejuvenation (97). Pre-existing clones provide a rapid antitumor response, while new clonotypes, undetectable before treatment, emerge post-ICB therapy (98). This dynamic relies heavily on continuous trafficking (99,100) between the lymphatic system and blood circulation. Notably, blocking lymphocyte egress from lymph nodes with the sphingosine-1-phosphate receptor inhibitor FTY720 abolishes ICB efficacy (101), underscoring the critical role of the peripheral immune system in T-cell activation and expansion.

Lymphoid structures

Peripheral lymphoid structures are central to adaptive immunity, serving as hubs where antigens, antigen-presenting cells (APCs) and lymphocytes interact within specialized microenvironments (Fig. 3). These interactions occur in secondary lymphoid organs (SLOs), such as tumor-draining lymph nodes (tdLNs), and in tertiary lymphoid structures (TLSs), which form in response to chronic inflammation (102). Both SLOs and TLSs facilitate antigen-specific T-cell activation, playing pivotal roles in antitumor immunity (103).

tdLNs

tdLNs are critical for antitumor immunity (104) and serve as both initial metastatic sites and primary locations for T-cell activation. The presence of these lymph nodes correlates with ICB efficacy (105), systemic immune responses (106) and improved survival (107). Mechanistically, tdLNs (108) support antitumor immunity through two pathways: De novo T-cell generation and maintenance of pre-existing T cells. Specialized dendritic cells (DCs) transport tumor antigens from the TME to tdLNs (109), where resident DCs activate naive T cells, initiating a cycle of T-cell trafficking between the lymph nodes and the TME (110). tdLNs harbor TCF1+ T cells with superior functionality compared to TILs (111,112). These cells, maintained by conventional type 1 dendritic cells (113,114), expand upon ICB treatment (62,115) and are key drivers of T-cell infiltration into the TME (105).

However, tdLNs can also foster immunosuppressive environments, with PD-L1+ (116) myeloid cells and regulatory T cells (Tregs) dampening T-cell responses. Tumor metastasis to tdLNs exacerbates this suppression, promoting distant metastasis and poor prognosis (117,118). Optimizing tdLN function presents a promising avenue for next-generation immunotherapies.

TLSs

TLSs are organized immune cell aggregates that form in chronic inflammatory conditions, including cancer (119-121). Structurally resembling SLOs, TLSs support de novo and ongoing immune responses, promoting T-cell activation and B-cell-mediated antibody production (122). Their presence is associated with favorable outcomes across multiple cancer types, including breast (123), colorectal (124) and pancreatic (125,126) cancer.

TLS formation is driven by chemokines such as CXCL13, and their maturity, ranging from early lymphocyte aggregation to fully developed germinal centers, impacts their prognostic value (127-129). Mature TLSs are linked to better outcomes, while immature TLSs may correlate with a poor prognosis in certain cancer types, such as lung and breast cancer (130-132). Emerging therapeutic strategies aim to enhance TLS formation (133-136) and maturation while mitigating tumor-promoting inflammation.

The heterogeneity in TLS maturation, categorized into early (lymphocyte aggregation), primary (follicular dendritic cells without mature DCs) and secondary stages [active germinal centers (GCs)] (137), is evident across cancers and assessed via markers such as CD138+ plasma cells (138) and CD23+ GC B cells (139). For example, esophageal cancer shows 26.9% TLS-negative, 30.2% immature and 42.9% mature cases, with mature TLSs featuring proliferative/memory B cells, plasma cells and CD4+ Th17 cells (140). Liver cancer exhibits 53% TLS-negative cases, 26% lymphoid aggregates, 16% primary and 5% secondary follicles, with the latter two linked to reduced recurrence risk (141). In squamous lung cancer, mature TLSs with GCs predict prognosis unless diminished by corticosteroids during neoadjuvant chemotherapy (128,142). Similarly, melanoma highlights B-cell markers (activation-induced cytidine deaminase and CD21) for mature TLSs and prognostic insights (143), while colorectal cancer emphasizes the significance of follicular dendritic cells and mature B cells (144).

However, TLSs may correlate with poor outcomes in some cancer types (145,146), such as renal cell carcinoma and pancreatic ductal adenocarcinoma, potentially due to TLS-associated Tregs (147), IL-10-secreting B cells (148), immune complexes, or tumor-promoting inflammation and angiogenesis (149,150). Emerging therapies aim to foster TLS neogenesis/maturation while mitigating inflammatory TMEs using cytokines, antibodies, immune checkpoint inhibitors, cellular therapies and synthetic scaffolds (151).

Lymphocytes in circulation
Total lymphocytes

The peripheral blood reflects systemic immune status, with lymphocyte composition serving as a key biomarker for antitumor immunity. Baseline lymphopenia, observed in various cancer types (152), such as metastatic breast cancer, correlates with disease burden and treatment outcomes. While systemic chemotherapy often suppresses peripheral immunity, localized treatments can preserve immune function and synergize with ICB (153). Post-treatment lymphocyte counts are predictive of ICB response, with higher levels correlating with improved outcomes in melanoma, lung cancer and renal cell carcinoma (RCC) (154). Relative lymphocyte count demonstrates significant prognostic value in melanoma (155,156), non-small cell lung cancer and RCC (157).

TCRs

Circulating TCR diversity is a critical determinant of antitumor immunity (158). Reduced TCR diversity is linked to aggressive disease and a poor prognosis (159,160), while dynamic TCR remodeling occurs in response to therapy (161-163). Post-treatment TCR expansion, particularly in CD8+ T cells, predicts ICB efficacy (164,165). However, high pretreatment TCR clonality may indicate poor outcomes in breast and lung cancer (163,166). The presence of polyclonal effector CD8+ T cells responding to a limited number of immunodominant mutations, with individual neoantigens corresponding to specific effector T-cell clones, appears fundamental to ICB response (167). TCR diversification, particularly of CD4+ blood T-cell clones prior to ICB administration, confers therapeutic benefit in lung cancer (168,169), RCC (170) and urothelial cancer (171). TCR profiles also correlate with immune-related adverse events (irAEs) (172,173).

T-cell phenotype

The functional status of circulating T cells significantly impacts cancer immunity (174). Memory T cells, particularly central memory T cell (Tcm) and effector memory T cell (Tem) subsets, are associated with favorable outcomes, while Tregs and Tex-term often indicate a poor prognosis (175-177). Treatment-induced activation of cytotoxic T cells and interferon-γ (IFNγ) production (174) are key markers of ICB response. Elevated PD1+/CD8+ T cells and natural killer (NK) cells (178) predict better outcomes, whereas increased Tregs and naive T cells correlate with a poor prognosis.

The composition of lymphocyte subsets reflects distinct functional signatures defined by specific phenotypic markers, with memory T-cell phenotypes generally linked to favorable clinical outcomes. Key prognostic indicators include the Tcm/effector T cell ratio (179), CD8+ Tem marked as C-C chemokine receptor type 7 (CCR7)/CD45RA/CD27+/CD28+ (180,181), and CD4+ memory T cells characterized by positivity for CD45RO/CCR7 and the absence of CD62L (182). Elevated baseline CD274 expression on T cells is also a significant prognostic marker (183). CD4+ T-cell subsets include central memory (CD45RA/CD62L+), effector memory (CD45RA/CD62L) and non-senescent (CD57) cells (184). Baseline PD1+ T cells and activated NK cells indicate antitumor immunity (185), while CD25+/FOXP3+/CD4+ Tregs are negative prognostic factors (182,184), with PD1 expression on Tregs linked to susceptibility to ICB-mediated activation and potential tumor hyperprogression (186). Tex-term (PD1+/T-cell immunoreceptor with Ig and ITIM domains+/LAG3+ and CD28/CD57+/killer cell lectin-like receptor subfamily G member 1+) (180,187,188) and PDL1+ NK cells further highlight the complexity of immune dynamics in the TME (157).

Treatment-induced immune changes, particularly early T-cell activation and proliferation marked by Ki-67 expression in PD1+/CD8+ Tex (189-193), cytotoxic activity, IFNγ activation and effector memory phenotypes (194,195), are critical indicators of ICB efficacy. A higher PD1+/CD8+ to PD1+/CD4+ T-cell ratio (173,196) and increased NK cell populations (197) correlate with favorable outcomes, whereas Treg induction predicts a poor prognosis (198). CD4+ Tcm (CD27+/FAS/CD45RA/CCR7+) play a vital role (199), and post-treatment T cell immunoglobulin and mucin domain-containing protein 3-positive Tex show tumor-specific prognostic value, being unfavorable in NSCLC and RCC, but favorable in esophageal squamous cell carcinoma (157,200). These findings suggest their utility as biomarkers for immune activation rather than as direct effectors. In hepatocellular carcinoma, an inflammatory signature involving PDL1, CD8A, LAG3 and signal transducer and activator of transcription 1 correlates with better outcomes, while macrophage-associated gene patterns show no prognostic significance (201). A composite biomarker panel, including mitochondrial activation (peroxisome proliferator-activated receptor γ coactivator 1 and reactive oxygen species) and specific T-cell populations (CD8+/PD-1hi and CD4+ T cells), has demonstrated high predictive accuracy for PD-1 blockade therapy outcomes (area under the curve, 0.96) (202).

Response and irAEs

irAEs are associated with enhanced therapeutic responses and prolonged survival times (203,204) in ICB-treated patients. Specific immune cell subsets, such as CXCR3+/CD8+ Tem and CD11c+ APCs, are linked to both efficacy and toxicity (205,206). Elevated Th17 (207) and Th1 (208) cells, coupled with reduced Treg levels, predict irAEs, while CTLA4 (209) insufficiency exacerbates these effects. Biomarkers such as post-treatment TCR clonality and peripheral immune profiles hold promise for optimizing treatment efficacy while minimizing irAEs (206,210).

T-cell therapeutics

Systemic inflammation

Systemic inflammation plays a pivotal role in regulating T-cell responses during immune defense against malignancies. Pro-inflammatory mediators, such as IL-1β, TNF-α and IL-6, create an inflammatory environment that facilitates APC-mediated T-cell activation and expansion. While this process is essential for initiating immune responses against infections and cancer, chronic inflammation can lead to T-cell exhaustion (211), marked by reduced effector functions and increased expression of inhibitory receptors such as PD-1 and CTLA-4.

Therapeutic strategies targeting systemic inflammation aim to enhance T-cell functionality while preventing exhaustion. Approaches include use of cytokine inhibitors (e.g., IL-6 or TNF-α antagonists) to mitigate excessive inflammation and checkpoint inhibitors to restore T-cell activity (212). Emerging therapies such as selective adjuvants that activate Toll-like receptors and metabolic interventions targeting T cell-specific pathways offer promising avenues to sustain T-cell function without inducing chronic inflammation, particularly in cancer immunotherapy contexts (213).

PD-1 blockade therapy

PD-1 blockade therapy has transformed cancer treatment by reinvigorating T cell-mediated antitumor immunity. PD-1, an immune checkpoint expressed on T cells, suppresses T-cell activation to maintain self-tolerance, but is exploited by tumors for immune evasion via the PD-1/PD-L1 axis (214). Monoclonal antibodies targeting this pathway, such as nivolumab and pembrolizumab, have achieved marked clinical success across various cancer types (215-217), such as non-small cell lung cancer, head and neck squamous cell carcinoma, and melanoma. Although these therapies provide durable responses and improved survival, they can cause irAEs, necessitating careful management (218,219). Combining PD-1 inhibitors with other immunotherapies or conventional treatments has shown promise in overcoming resistance and achieving synergistic effects (220-223).

Vaccine technologies

Advances in vaccine technologies have significantly enhanced T cell-mediated antitumor immunity. Cancer vaccines targeting tumor-associated antigens stimulate specific T-cell responses by mimicking natural infections (224-227). The addition of adjuvants, such as CpG oligodeoxynucleotides, further amplifies innate immune responses, supporting T-cell activation and proliferation (228-231). Combining cancer vaccines with checkpoint inhibitors has demonstrated synergistic effects, improving T-cell responses and tumor clearance (225,232,233). Personalized cancer vaccines, tailored to individual tumor antigenic profiles, represent a promising breakthrough by generating broad-spectrum T-cell responses while minimizing immune escape. These developments mark a significant step forward in precision immunotherapy (234).

mRNA-based therapies

mRNA-based therapies have emerged as a powerful tool in cancer immunotherapy, leveraging the ability of mRNA to encode tumor-specific antigens and stimulate targeted immune responses. Advances in delivery systems, such as lipid nanoparticles, have improved mRNA stability and protection against degradation (235-237). Structural modifications, including nucleoside alterations, enhance translation efficiency while minimizing immunogenicity (238-241). Co-delivery of immunomodulatory molecules encoded by mRNA further amplifies T-cell responses and counters immunosuppression (242). Clinical trials, particularly in melanoma, have demonstrated the efficacy of personalized mRNA vaccines in eliciting robust T-cell responses, underscoring their potential in cancer treatment (243-245).

Nano delivery systems

Lymphoid tissue-targeted nano delivery systems offer an innovative approach to enhancing T-cell functionality in cancer immunotherapy. Engineered nanoparticles, modified with ligands targeting lymphoid tissue receptors (e.g., CCR7 and CXCR5), enable precise delivery of immunomodulatory agents (246-249). Encapsulation of cytokines, such as IL-2 and IL-12 (250-253), within these nanoparticles enhances local T-cell responses while minimizing systemic side effects. Recent advancements include delivering checkpoint inhibitors directly to lymphoid tissues, addressing T-cell exhaustion in the TME (254-257). By concentrating therapeutic agents at desired sites, these systems improve treatment efficacy and reduce adverse effects, representing a promising direction for cancer immunotherapy.

Oncolytic viruses (Ovs)

OVs present a novel strategy to enhance T cell-mediated antitumor responses by selectively infecting and lysing cancer cells. This process triggers pro-inflammatory responses that activate adaptive immunity while sparing healthy tissue (258). OVs can also be engineered to express immunomodulatory genes, such as IL-12 and granulocyte-macrophage colony-stimulating factor, which enhance T-cell recruitment and activation within the TME (259-262). Combining OVs with other immunotherapies, particularly checkpoint inhibitors, has shown synergistic effects in overcoming tumor-induced immunosuppression (263). This dual mechanism, namely direct tumor lysis and immune activation, positions OVs as a promising component of combination immunotherapy strategies.

Perspective

The present review underscores the intricate interplay between local and systemic factors in T cell-mediated antitumor immunity. Within the TME, T cells face challenges such as physical exclusion, functional exhaustion and immunosuppression. The distinction between Tex-prog and Tex-term has emerged as a key factor influencing immunotherapy outcomes. Beyond the tumor site, systemic immune responses mediated by lymphoid structures and circulation are critical for sustaining effective antitumor immunity.

Recent advancements in T-cell biology have paved the way for novel therapeutic strategies, including mRNA-based therapies, targeted nano delivery systems and engineered oncolytic viruses. These innovations, coupled with improved biomarkers, such as TCR repertoire analysis and circulating T-cell phenotyping, enable more precise patient stratification and treatment optimization.

Looking ahead, several challenges remain. Balancing therapeutic efficacy with irAEs requires deeper insights into the molecular mechanisms distinguishing beneficial from harmful immune responses. Additionally, the role of systemic immunity, particularly lymphoid structures and circulation, warrants further exploration. Combination therapies targeting both T cells and the TME hold significant potential but need systematic evaluation. Finally, personalized immunotherapy, incorporating patient-specific factors, such as tumor antigens and immune status, promises to enhance treatment outcomes. Future clinical trials should consider stratifying patients according to Tex-prog and Tex-term profiles to optimize immunotherapy outcomes. Additionally, therapeutic interventions that promote the formation and maturation of TLSs in selected tumor types warrant systematic evaluation.

In conclusion, the rapid evolution of T cell-based cancer immunotherapy offers exciting opportunities to overcome current limitations. By advancing our understanding of immune dynamics and leveraging emerging technologies, more effective and personalized treatment strategies can be developed, ultimately expanding the benefits of immunotherapy to a broader patient population.

Availability of data and materials

Not applicable.

Authors' contributions

XL, JH and CL wrote the daft. YZ and WW reviewed and revised the manuscript. All authors have read and approved the final manuscript. Data authentication is not applicable.

Ethics approval and consent to participate

Not applicable.

Patient consent for publication

Not applicable.

Competing interests

The authors declare that they have no competing interests.

Acknowledgements

Not applicable.

Funding

This study was supported by grants from the National High Level Hospital Clinical Research Funding (grant no. 2022-PUMCH-B-039 and grant no. 2025-PUMCH-D-001), and the National College Student Innovation and Entrepreneurship Training Program (grant nos. 202110023010, 2022zglc06080 and 2023zglc06022).

References

1 

Anagnostou V, Niknafs N, Marrone K, Bruhm DC, White JR, Naidoo J, Hummelink K, Monkhorst K, Lalezari F, Lanis M, et al: Multimodal genomic features predict outcome of immune checkpoint blockade in non-small-cell lung cancer. Nat Cancer. 1:99–111. 2020. View Article : Google Scholar

2 

Wu B, Zhang B, Li B, Wu H and Jiang M: Cold and hot tumors: From molecular mechanisms to targeted therapy. Signal Transduct Target Ther. 9:2742024. View Article : Google Scholar :

3 

Yu Y, Zeng D, Ou Q, Liu S, Li A, Chen Y, Lin D, Gao Q, Zhou H, Liao W and Yao H: Association of survival and immune-related biomarkers with immunotherapy in patients with non-small cell lung cancer: A meta-analysis and individual patient-level analysis. JAMA Netw Open. 2:e1968792019. View Article : Google Scholar

4 

Spitzer MH, Carmi Y, Reticker-Flynn NE, Kwek SS, Madhireddy D, Martins MM, Gherardini PF, Prestwood TR, Chabon J, Bendall SC, et al: Systemic immunity is required for effective cancer immunotherapy. Cell. 168:487–502.e15. 2017. View Article : Google Scholar

5 

Tselikas L, Dardenne A, de Baere T, Faron M, Ammari S, Farhane S, Suzzoni S, Danlos FX, Raoult T, Susini S, et al: Feasibility, safety and efficacy of human intra-tumoral immuno-therapy. Gustave Roussy's initial experience with its first 100 patients. Eur J Cancer. 172:1–12. 2022. View Article : Google Scholar

6 

Spranger S: Mechanisms of tumor escape in the context of the T-cell-inflamed and the non-T-cell-inflamed tumor microenvironment. Int Immunol. 28:383–391. 2016. View Article : Google Scholar :

7 

Sadeghi Rad H, Monkman J, Warkiani ME, Ladwa R, O'Byrne K, Rezaei N and Kulasinghe A: Understanding the tumor microenvironment for effective immunotherapy. Med Res Rev. 41:1474–1498. 2021. View Article : Google Scholar :

8 

Casalegno Garduño R, Spitschak A, Pannek T and Pützer BM: CD8+ T cell subsets as biomarkers for predicting checkpoint therapy outcomes in cancer immunotherapy. Biomedicines. 13:9302025. View Article : Google Scholar

9 

Loi S, Adams S, Schmid P, Cortés J, Cescon DW, Winer EP, Toppmeyer DL, Rugo HS, De Laurentiis M, Nanda R, et al: LBA13-Relationship between tumor infiltrating lymphocyte (TIL) levels and response to pembrolizumab (pembro) in metastatic triple-negative breast cancer (mTNBC): Results from KEYNOTE-086. Ann Oncol. 28(Suppl 5): v6082017. View Article : Google Scholar

10 

Hegde PS and Chen DS: Top 10 challenges in cancer immunotherapy. Immunity. 52:17–35. 2020. View Article : Google Scholar : PubMed/NCBI

11 

Hugo W, Zaretsky JM, Sun L, Song C, Moreno BH, Hu-Lieskovan S, Berent-Maoz B, Pang J, Chmielowski B, Cherry G, et al: Genomic and transcriptomic features of response to Anti-PD-1 therapy in metastatic melanoma. Cell. 165:35–44. 2016. View Article : Google Scholar : PubMed/NCBI

12 

Mariathasan S, Turley SJ, Nickles D, Castiglioni A, Yuen K, Wang Y, Kadel EE III, Koeppen H, Astarita JL, Cubas R, et al: TGFβ attenuates tumour response to PD-L1 blockade by contributing to exclusion of T cells. Nature. 554:544–548. 2018. View Article : Google Scholar

13 

Cascio S, Chandler C, Zhang L, Sinno S, Gao B, Onkar S, Bruno TC, Vignali DAA, Mahdi H, Osmanbeyoglu HU, et al: Cancer-associated MSC drive tumor immune exclusion and resistance to immunotherapy, which can be overcome by Hedgehog inhibition. Sci Adv. 7:eabi57902021. View Article : Google Scholar : PubMed/NCBI

14 

Grout JA, Sirven P, Leader AM, Maskey S, Hector E, Puisieux I, Steffan F, Cheng E, Tung N, Maurin M, et al: Spatial positioning and matrix programs of cancer-associated fibroblasts promote T-cell exclusion in human lung tumors. Cancer Discov. 12:2606–2625. 2022. View Article : Google Scholar : PubMed/NCBI

15 

Elyada E, Bolisetty M, Laise P, Flynn WF, Courtois ET, Burkhart RA, Teinor JA, Belleau P, Biffi G, Lucito MS, et al: Cross-species single-cell analysis of pancreatic ductal adenocarcinoma reveals antigen-presenting cancer-associated fibroblasts. Cancer Discov. 9:1102–1123. 2019. View Article : Google Scholar : PubMed/NCBI

16 

Griffioen AW, Damen CA, Blijham GH and Groenewegen G: Tumor angiogenesis is accompanied by a decreased inflammatory response of tumor-associated endothelium. Blood. 88:667–673. 1996. View Article : Google Scholar

17 

Nagl L, Horvath L, Pircher A and Wolf D: Tumor endothelial cells (TECs) as potential immune directors of the tumor microenvironment-new findings and future perspectives. Front Cell Dev Biol. 8:7662020. View Article : Google Scholar

18 

Sahu A, Kose K, Kraehenbuehl L, Byers C, Holland A, Tembo T, Santella A, Alfonso A, Li M, Cordova M, et al: In vivo tumor immune microenvironment phenotypes correlate with inflammation and vasculature to predict immunotherapy response. Nat Commun. 13:53122022. View Article : Google Scholar

19 

Subramanian M, Kabir AU, Barisas D, Krchma K and Choi K: Conserved angio-immune subtypes of the tumor microenvironment predict response to immune checkpoint blockade therapy. Cell Rep Med. 4:1008962023. View Article : Google Scholar : PubMed/NCBI

20 

Steele MM, Jaiswal A, Delclaux I, Dryg ID, Murugan D, Femel J, Son S, du Bois H, Hill C, Leachman SA, et al: T cell egress via lymphatic vessels is tuned by antigen encounter and limits tumor control. Nat Immunol. 24:664–675. 2023. View Article : Google Scholar : PubMed/NCBI

21 

Huang CX, Lao XM, Wang XY, Ren YZ, Lu YT, Shi W, Wang YZ, Wu CY, Xu L, Chen MS, et al: Pericancerous cross-presentation to cytotoxic T lymphocytes impairs immunotherapeutic efficacy in hepatocellular carcinoma. Cancer Cell. 42:2082–2097.e10. 2024. View Article : Google Scholar

22 

Marabelle A, Fakih M, Lopez J, Shah M, Shapira-Frommer R, Nakagawa K, Chung HC, Kindler HL, Lopez-Martin JA, Miller WH Jr, et al: Association of tumour mutational burden with outcomes in patients with advanced solid tumours treated with pembrolizumab: Prospective biomarker analysis of the multicohort, open-label, phase 2 KEYNOTE-158 study. Lancet Oncol. 21:1353–1365. 2020. View Article : Google Scholar : PubMed/NCBI

23 

Cristescu R, Aurora-Garg D, Albright A, Xu L, Liu XQ, Loboda A, Lang L, Jin F, Rubin EH, Snyder A and Lunceford J: Tumor mutational burden predicts the efficacy of pembrolizumab monotherapy: A pan-tumor retrospective analysis of participants with advanced solid tumors. J Immunother Cancer. 10:e0030912022. View Article : Google Scholar : PubMed/NCBI

24 

Ricciuti B, Wang X, Alessi JV, Rizvi H, Mahadevan NR, Li YY, Polio A, Lindsay J, Umeton R, Sinha R, et al: Association of high tumor mutation burden in non-small cell lung cancers with increased immune infiltration and improved clinical outcomes of PD-L1 blockade across PD-L1 expression levels. JAMA Oncol. 8:1160–1168. 2022. View Article : Google Scholar : PubMed/NCBI

25 

Baharom F, Ramirez-Valdez RA, Khalilnezhad A, Khalilnezhad S, Dillon M, Hermans D, Fussell S, Tobin KKS, Dutertre CA, Lynn GM, et al: Systemic vaccination induces CD8+ T cells and remodels the tumor microenvironment. Cell. 185:4317–4332.e15. 2022. View Article : Google Scholar

26 

Zheng M: Tumor mutation burden for predicting immune checkpoint blockade response: The more, the better. J Immunother Cancer. 10:e0030872022. View Article : Google Scholar

27 

McGrail DJ, Pilié PG, Rashid NU, Voorwerk L, Slagter M, Kok M, Jonasch E, Khasraw M, Heimberger AB, Lim B, et al: High tumor mutation burden fails to predict immune checkpoint blockade response across all cancer types. Ann Oncol. 32:661–672. 2021. View Article : Google Scholar : PubMed/NCBI

28 

Niknafs N, Balan A, Cherry C, Hummelink K, Monkhorst K, Shao XM, Belcaid Z, Marrone KA, Murray J, Smith KN, et al: Persistent mutation burden drives sustained anti-tumor immune responses. Nat Med. 29:440–449. 2023. View Article : Google Scholar : PubMed/NCBI

29 

Valpione S, Mundra PA, Galvani E, Campana LG, Lorigan P, De Rosa F, Gupta A, Weightman J, Mills S, Dhomen N and Marais R: The T cell receptor repertoire of tumor infiltrating T cells is predictive and prognostic for cancer survival. Nat Commun. 12:40982021. View Article : Google Scholar : PubMed/NCBI

30 

Meier SL, Satpathy AT and Wells DK: Bystander T cells in cancer immunology and therapy. Nat Cancer. 3:143–155. 2022. View Article : Google Scholar : PubMed/NCBI

31 

Simoni Y, Becht E, Fehlings M, Loh CY, Koo SL, Teng KWW, Yeong JPS, Nahar R, Zhang T, Kared H, et al: Bystander CD8+ T cells are abundant and phenotypically distinct in human tumour infiltrates. Nature. 557:575–579. 2018. View Article : Google Scholar : PubMed/NCBI

32 

Chen X, Zhao J, Yue S, Li Z, Duan X, Lin Y, Yang Y, He J, Gao L, Pan Z, et al: An oncolytic virus delivering tumor-irrelevant bystander T cell epitopes induces anti-tumor immunity and potentiates cancer immunotherapy. Nat Cancer. 5:1063–1081. 2024. View Article : Google Scholar : PubMed/NCBI

33 

Lowery FJ, Krishna S, Yossef R, Parikh NB, Chatani PD, Zacharakis N, Parkhurst MR, Levin N, Sindiri S, Sachs A, et al: Molecular signatures of antitumor neoantigen-reactive T cells from metastatic human cancers. Science. 375:877–884. 2022. View Article : Google Scholar : PubMed/NCBI

34 

Meng Z, Rodriguez Ehrenfried A, Tan CL, Steffens LK, Kehm H, Zens S, Lauenstein C, Paul A, Schwab M, Förster JD, et al: Transcriptome-based identification of tumor-reactive and bystander CD8+ T cell receptor clonotypes in human pancreatic cancer. Sci Transl Med. 15:eadh95622023. View Article : Google Scholar

35 

Kortekaas KE, Santegoets SJ, Sturm G, Ehsan I, van Egmond SL, Finotello F, Rajanoski Z, Welters MJP, van Poelgeest MIE and van der Burg SH: CD39 identifies the CD4+ tumor-specific T-cell population in human cancer. Cancer Immunol Res. 8:1311–1321. 2020. View Article : Google Scholar

36 

Duhen T, Duhen R, Montler R, Moses J, Moudgil T, de Miranda NF, Goodall CP, Blair TC, Fox BA, McDermott JE, et al: Co-expression of CD39 and CD103 identifies tumor-reactive CD8 T cells in human solid tumors. Nat Commun. 9:27242018. View Article : Google Scholar

37 

Qiao M, Zhou F, Liu X, Jiang T, Wang H, Jia Y, Li X, Zhao C, Cheng L, Chen X, et al: Interleukin-10 induces expression of CD39 on CD8+T cells to potentiate anti-PD1 efficacy in EGFR-mutated non-small cell lung cancer. J Immunother Cancer. 10:e0054362022. View Article : Google Scholar : PubMed/NCBI

38 

Webb JR, Milne K and Nelson BH: PD-1 and CD103 are widely coexpressed on prognostically favorable intraepithelial CD8 T cells in human ovarian cancer. Cancer Immunol Res. 3:926–935. 2015. View Article : Google Scholar : PubMed/NCBI

39 

Corgnac S, Malenica I, Mezquita L, Auclin E, Voilin E, Kacher J, Halse H, Grynszpan L, Signolle N, Dayris T, et al: CD103+CD8+ TRM cells accumulate in tumors of anti-PD-1-responder lung cancer patients and are tumor-reactive lymphocytes enriched with Tc17. Cell Rep Med. 1:1001272020. View Article : Google Scholar

40 

Wang Z, Ahmed S, Labib M, Wang H, Wu L, Bavaghar-Zaeimi F, Shokri N, Blanco S, Karim S, Czarnecka-Kujawa K, et al: Isolation of tumour-reactive lymphocytes from peripheral blood via microfluidic immunomagnetic cell sorting. Nat Biomed Eng. 7:1188–1203. 2023. View Article : Google Scholar : PubMed/NCBI

41 

Gallerano D, Ciminati S, Grimaldi A, Piconese S, Cammarata I, Focaccetti C, Pacella I, Accapezzato D, Lancellotti F, Sacco L, et al: Genetically driven CD39 expression shapes human tumor-infiltrating CD8+ T-cell functions. Int J Cancer. 147:2597–2610. 2020. View Article : Google Scholar : PubMed/NCBI

42 

Laumont CM, Wouters MCA, Smazynski J, Gierc NS, Chavez EA, Chong LC, Thornton S, Milne K, Webb JR, Steidl C and Nelson BH: Single-cell profiles and prognostic impact of tumor-infiltrating lymphocytes coexpressing CD39, CD103, and PD-1 in ovarian cancer. Clin Cancer Res. 27:4089–4100. 2021. View Article : Google Scholar : PubMed/NCBI

43 

He J, Xiong X, Yang H, Li D, Liu X, Li S, Liao S, Chen S, Wen X, Yu K, et al: Defined tumor antigen-specific T cells potentiate personalized TCR-T cell therapy and prediction of immunotherapy response. Cell Res. 32:530–542. 2022. View Article : Google Scholar

44 

Liu B, Zhang Y, Wang D, Hu X and Zhang Z: Single-cell meta-analyses reveal responses of tumor-reactive CXCL13+ T cells to immune-checkpoint blockade. Nat Cancer. 3:1123–1136. 2022. View Article : Google Scholar

45 

Hanada KI, Zhao C, Gil-Hoyos R, Gartner JJ, Chow-Parmer C, Lowery FJ, Krishna S, Prickett TD, Kivitz S, Parkhurst MR, et al: A phenotypic signature that identifies neoantigen-reactive T cells in fresh human lung cancers. Cancer Cell. 40:479–493.e6. 2022. View Article : Google Scholar : PubMed/NCBI

46 

Dai S, Zeng H, Liu Z, Jin K, Jiang W, Wang Z, Lin Z, Xiong Y, Wang J, Chang Y, et al: Intratumoral CXCL13+CD8+T cell infiltration determines poor clinical outcomes and immunoevasive contexture in patients with clear cell renal cell carcinoma. J Immunother Cancer. 9:e0018232021. View Article : Google Scholar

47 

Aoki T, Chong LC, Takata K, Milne K, Marshall A, Chavez EA, Miyata-Takata T, Ben-Neriah S, Unrau D, Telenius A, et al: Single-cell profiling reveals the importance of CXCL13/CXCR5 axis biology in lymphocyte-rich classic Hodgkin lymphoma. Proc Natl Acad Sci USA. 118:e21058221182021. View Article : Google Scholar :

48 

Eiva MA, Omran DK, Chacon JA and Powell DJ Jr: Systematic analysis of CD39, CD103, CD137, and PD-1 as biomarkers for naturally occurring tumor antigen-specific TILs. Eur J Immunol. 52:96–108. 2022. View Article : Google Scholar :

49 

Parkhurst M, Gros A, Pasetto A, Prickett T, Crystal JS, Robbins P and Rosenberg SA: Isolation of T-cell receptors specifically reactive with mutated tumor-associated antigens from tumor-infiltrating lymphocytes based on CD137 expression. Clin Cancer Res. 23:2491–2505. 2017. View Article : Google Scholar

50 

Yost KE, Satpathy AT, Wells DK, Qi Y, Wang C, Kageyama R, McNamara KL, Granja JM, Sarin KY, Brown RA, et al: Clonal replacement of tumor-specific T cells following PD-1 blockade. Nat Med. 25:1251–1259. 2019. View Article : Google Scholar

51 

Tonnerre P, Wolski D, Subudhi S, Aljabban J, Hoogeveen RC, Damasio M, Drescher HK, Bartsch LM, Tully DC, Sen DR, et al: Differentiation of exhausted CD8+ T cells after termination of chronic antigen stimulation stops short of achieving functional T cell memory. Nat Immunol. 22:1030–1041. 2021. View Article : Google Scholar : PubMed/NCBI

52 

Watowich MB, Gilbert MR and Larion M: T cell exhaustion in malignant gliomas. Trends Cancer. 9:270–292. 2023. View Article : Google Scholar : PubMed/NCBI

53 

Beltra JC, Manne S, Abdel-Hakeem MS, Kurachi M, Giles JR, Chen Z, Casella V, Ngiow SF, Khan O, Huang YJ, et al: Developmental relationships of four exhausted CD8+ T cell subsets reveals underlying transcriptional and epigenetic landscape control mechanisms. Immunity. 52:825–841.e8. 2020. View Article : Google Scholar

54 

Sen DR, Kaminski J, Barnitz RA, Kurachi M, Gerdemann U, Yates KB, Tsao HW, Godec J, LaFleur MW, Brown FD, et al: The epigenetic landscape of T cell exhaustion. Science. 354:1165–1169. 2016. View Article : Google Scholar : PubMed/NCBI

55 

Zhang Z, Chen L, Chen H, Zhao J, Li K, Sun J and Zhou M: Pan-cancer landscape of T-cell exhaustion heterogeneity within the tumor microenvironment revealed a progressive roadmap of hierarchical dysfunction associated with prognosis and therapeutic efficacy. EBioMedicine. 83:1042072022. View Article : Google Scholar : PubMed/NCBI

56 

Wu Z, Yoshikawa T, Inoue S, Ito Y, Kasuya H, Nakashima T, Zhang H, Kotaka S, Hosoda W, Suzuki S and Kagoya Y: CD83 expression characterizes precursor exhausted T cell population. Commun Biol. 6:2582023. View Article : Google Scholar : PubMed/NCBI

57 

Utzschneider DT, Charmoy M, Chennupati V, Pousse L, Ferreira DP, Calderon-Copete S, Danilo M, Alfei F, Hofmann M, Wieland D, et al: T cell factor 1-expressing memory-like CD8(+) T cells sustain the immune response to chronic viral infections. Immunity. 45:415–427. 2016. View Article : Google Scholar

58 

Kim CG, Kim G, Kim KH, Park S, Shin S, Yeo D, Shim HS, Yoon HI, Park SY, Ha SJ and Kim HR: Distinct exhaustion features of T lymphocytes shape the tumor-immune microenvironment with therapeutic implication in patients with non-small-cell lung cancer. J Immunother Cancer. 9:e0027802021. View Article : Google Scholar : PubMed/NCBI

59 

Wieland D, Kemming J, Schuch A, Emmerich F, Knolle P, Neumann-Haefelin C, Held W, Zehn D, Hofmann M and Thimme R: TCF1+ hepatitis C virus-specific CD8+ T cells are maintained after cessation of chronic antigen stimulation. Nat Commun. 8:150502017. View Article : Google Scholar

60 

Jadhav RR, Im SJ, Hu B, Hashimoto M, Li P, Lin JX, Leonard WJ, Greenleaf WJ, Ahmed R and Goronzy JJ: Epigenetic signature of PD-1+ TCF1+ CD8 T cells that act as resource cells during chronic viral infection and respond to PD-1 blockade. Proc Natl Acad Sci USA. 116:14113–14118. 2019. View Article : Google Scholar : PubMed/NCBI

61 

Li X, Li Y, Dong L, Chang Y, Zhang X, Wang C, Chen M, Bo X, Chen H, Han W and Nie J: Decitabine priming increases anti-PD-1 antitumor efficacy by promoting CD8+ progenitor exhausted T cell expansion in tumor models. J Clin Invest. 133:e1656732023. View Article : Google Scholar : PubMed/NCBI

62 

Nagasaki J, Inozume T, Sax N, Ariyasu R, Ishikawa M, Yamashita K, Kawazu M, Ueno T, Irie T, Tanji E, et al: PD-1 blockade therapy promotes infiltration of tumor-attacking exhausted T cell clonotypes. Cell Rep. 38:1103312022. View Article : Google Scholar

63 

Codarri Deak L, Nicolini V, Hashimoto M, Karagianni M, Schwalie PC, Lauener L, Varypataki EM, Richard M, Bommer E, Sam J, et al: PD-1-cis IL-2R agonism yields better effectors from stem-like CD8+ T cells. Nature. 610:161–172. 2022. View Article : Google Scholar : PubMed/NCBI

64 

Hashimoto M, Araki K, Cardenas MA, Li P, Jadhav RR, Kissick HT, Hudson WH, McGuire DJ, Obeng RC, Wieland A, et al: PD-1 combination therapy with IL-2 modifies CD8+ T cell exhaustion program. Nature. 610:173–181. 2022. View Article : Google Scholar :

65 

Ren Z, Zhang A, Sun Z, Liang Y, Ye J, Qiao J, Li B and Fu YX: Selective delivery of low-affinity IL-2 to PD-1+ T cells rejuvenates antitumor immunity with reduced toxicity. J Clin Invest. 132:e1536042022. View Article : Google Scholar

66 

Zehn D, Thimme R, Lugli E, de Almeida GP and Oxenius A: 'Stem-like' precursors are the fount to sustain persistent CD8+ T cell responses. Nat Immunol. 23:836–847. 2022. View Article : Google Scholar

67 

Miller BC, Sen DR, Al Abosy R, Bi K, Virkud YV, LaFleur MW, Yates KB, Lako A, Felt K, Naik GS, et al: Subsets of exhausted CD8+ T cells differentially mediate tumor control and respond to checkpoint blockade. Nat Immunol. 20:326–336. 2016. View Article : Google Scholar

68 

Im SJ, Hashimoto M, Gerner MY, Lee J, Kissick HT, Burger MC, Shan Q, Hale JS, Lee J, Nasti TH, et al: Defining CD8+ T cells that provide the proliferative burst after PD-1 therapy. Nature. 537:417–421. 2016. View Article : Google Scholar

69 

Siddiqui I, Schaeuble K, Chennupati V, Fuertes Marraco SA, Calderon-Copete S, Pais Ferreira D, Carmona SJ, Scarpellino L, Gfeller D, Pradervand S, et al: Intratumoral Tcf1+PD-1+CD8+ T cells with stem-like properties promote tumor control in response to vaccination and checkpoint blockade immunotherapy. Immunity. 50:195–211.e10. 2019. View Article : Google Scholar

70 

Tabanelli V, Melle F, Motta G, Mazzara S, Fabbri M, Agostinelli C, Calleri A, Del Corvo M, Fiori S, Lorenzini D, et al: The identification of TCF1+ progenitor exhausted T cells in THRLBCL may predict a better response to PD-1/PD-L1 blockade. Blood Adv. 6:4634–4644. 2022. View Article : Google Scholar :

71 

Zheng L, Qin S, Si W, Wang A, Xing B, Gao R, Ren X, Wang L, Wu X, Zhang J, et al: Pan-cancer single-cell landscape of tumor-infiltrating T cells. Science. 374:abe64742021. View Article : Google Scholar

72 

Alfei F, Kanev K, Hofmann M, Wu M, Ghoneim HE, Roelli P, Utzschneider DT, von Hoesslin M, Cullen JG, Fan Y, et al: TOX reinforces the phenotype and longevity of exhausted T cells in chronic viral infection. Nature. 571:265–269. 2019. View Article : Google Scholar : PubMed/NCBI

73 

Kim K, Park S, Park SY, Kim G, Park SM, Cho JW, Kim DH, Park YM, Koh YW, Kim HR, et al: Single-cell transcriptome analysis reveals TOX as a promoting factor for T cell exhaustion and a predictor for anti-PD-1 responses in human cancer. Genome Med. 12:222020. View Article : Google Scholar :

74 

Abdel-Hakeem MS, Manne S, Beltra JC, Stelekati E, Chen Z, Nzingha K, Ali MA, Johnson JL, Giles JR, Mathew D, et al: Epigenetic scarring of exhausted T cells hinders memory differentiation upon eliminating chronic antigenic stimulation. Nat Immunol. 22:1008–1019. 2021. View Article : Google Scholar :

75 

Pauken KE, Sammons MA, Odorizzi PM, Manne S, Godec J, Khan O, Drake AM, Chen Z, Sen DR, Kurachi M, et al: Epigenetic stability of exhausted T cells limits durability of reinvigoration by PD-1 blockade. Science. 354:1160–1165. 2016. View Article : Google Scholar

76 

Gupta PK, Godec J, Wolski D, Adland E, Yates K, Pauken KE, Cosgrove C, Ledderose C, Junger WG, Robson SC, et al: CD39 expression identifies terminally exhausted CD8+ T cells. PLoS Pathog. 11:e10051772015. View Article : Google Scholar : PubMed/NCBI

77 

Tinoco R, Neubert EN, Stairiker CJ, Henriquez ML and Bradley LM: PSGL-1 is a T cell intrinsic inhibitor that regulates effector and memory differentiation and responses during viral infection. Front Immunol. 12:6778242021. View Article : Google Scholar :

78 

Tinoco R, Carrette F, Barraza ML, Otero DC, Magaña J, Bosenberg MW, Swain SL and Bradley LM: PSGL-1 is an immune checkpoint regulator that promotes T cell exhaustion. Immunity. 44:1190–1203. 2016. View Article : Google Scholar

79 

Vignali PDA, DePeaux K, Watson MJ, Ye C, Ford BR, Lontos K, McGaa NK, Scharping NE, Menk AV, Robson SC, et al: Hypoxia drives CD39-dependent suppressor function in exhausted T cells to limit antitumor immunity. Nat Immunol. 24:267–279. 2023. View Article : Google Scholar

80 

Viramontes KM, Neubert EN, DeRogatis JM and Tinoco R: PD-1 immune checkpoint blockade and PSGL-1 inhibition synergize to reinvigorate exhausted T cells. Front Immunol. 13:8697682022. View Article : Google Scholar :

81 

Moesta AK, Li XY and Smyth MJ: Targeting CD39 in cancer. Nat Rev Immunol. 20:739–755. 2020. View Article : Google Scholar : PubMed/NCBI

82 

Tu E, McGlinchey K, Wang J, Martin P, Ching SL, Floc'h N, Kurasawa J, Starrett JH, Lazdun Y, Wetzel L, et al: Anti-PD-L1 and anti-CD73 combination therapy promotes T cell response to EGFR-mutated NSCLC. JCI Insight. 7:e1428432022. View Article : Google Scholar : PubMed/NCBI

83 

Ford BR and Poholek AC: Regulation and immunotherapeutic targeting of the epigenome in exhausted CD8 T cell responses. J Immunol. 210:869–879. 2023. View Article : Google Scholar

84 

Franco F, Jaccard A, Romero P, Yu YR and Ho PC: Metabolic and epigenetic regulation of T-cell exhaustion. Nat Metab. 2:1001–1012. 2020. View Article : Google Scholar : PubMed/NCBI

85 

Gabriel SS, Tsui C, Chisanga D, Weber F, Llano-León M, Gubser PM, Bartholin L, Souza-Fonseca-Guimaraes F, Huntington ND, Shi W, et al: Transforming growth factor-β-regulated mTOR activity preserves cellular metabolism to maintain long-term T cell responses in chronic infection. Immunity. 54:1698–1714.e5. 2021. View Article : Google Scholar

86 

Bengsch B, Johnson AL, Kurachi M, Odorizzi PM, Pauken KE, Attanasio J, Stelekati E, McLane LM, Paley MA, Delgoffe GM and Wherry EJ: Bioenergetic insufficiencies due to metabolic alterations regulated by the inhibitory receptor PD-1 are an early driver of CD8(+) T cell exhaustion. Immunity. 45:358–373. 2016. View Article : Google Scholar : PubMed/NCBI

87 

Guo Y, Xie YQ, Gao M, Zhao Y, Franco F, Wenes M, Siddiqui I, Bevilacqua A, Wang H, Yang H, et al: Metabolic reprogramming of terminally exhausted CD8+ T cells by IL-10 enhances anti-tumor immunity. Nat Immunol. 22:746–756. 2021. View Article : Google Scholar :

88 

Tsui C, Kretschmer L, Rapelius S, Gabriel SS, Chisanga D, Knöpper K, Utzschneider DT, Nüssing S, Liao Y, Mason T, et al: MYB orchestrates T cell exhaustion and response to checkpoint inhibition. Nature. 609:354–360. 2022. View Article : Google Scholar :

89 

Stelekati E, Chen Z, Manne S, Kurachi M, Ali MA, Lewy K, Cai Z, Nzingha K, McLane LM, Hope JL, et al: Long-term persistence of exhausted CD8 T cells in chronic infection is regulated by MicroRNA-155. Cell Rep. 23:2142–2156. 2018. View Article : Google Scholar : PubMed/NCBI

90 

Utzschneider DT, Gabriel SS, Chisanga D, Gloury R, Gubser PM, Vasanthakumar A, Shi W and Kallies A: Early precursor T cells establish and propagate T cell exhaustion in chronic infection. Nat Immun. 21:1256–1266. 2020. View Article : Google Scholar

91 

Man K, Gabriel SS, Liao Y, Gloury R, Preston S, Henstridge DC, Pellegrini M, Zehn D, Berberich-Siebelt F, Febbraio MA, et al: Transcription factor IRF4 promotes CD8+ T cell exhaustion and limits the development of memory-like T cells during chronic infection. Immunity. 47:1129–1141.e5. 2017. View Article : Google Scholar

92 

Seo H, González-Avalos E, Zhang W, Ramchandani P, Yang C, Lio CJ, Rao A and Hogan PG: BATF and IRF4 cooperate to counter exhaustion in tumor-infiltrating CAR T cells. Nat Immunol. 22:983–995. 2021. View Article : Google Scholar :

93 

Russ BE, Tsyganov K, Quon S, Yu B, Li J, Lee JKC, Olshansky M, He Z, Harrison PF, Barugahare A, et al: Active maintenance of CD8+ T cell naïvety through regulation of global genome architecture. bioRxiv: The preprint server for biology. 2023.

94 

Grusdat M, McIlwain DR, Xu HC, Pozdeev VI, Knievel J, Crome SQ, Robert-Tissot C, Dress RJ, Pandyra AA, Speiser DE, et al: IRF4 and BATF are critical for CD8+ T-cell function following infection with LCMV. Cell Death Differ. 21:1050–1060. 2014. View Article : Google Scholar : PubMed/NCBI

95 

Jain N, Zhao Z, Feucht J, Koche R, Iyer A, Dobrin A, Mansilla-Soto J, Yang J, Zhan Y, Lopez M, et al: TET2 guards against unchecked BATF3-induced CAR T cell expansion. Nature. 615:315–322. 2023. View Article : Google Scholar : PubMed/NCBI

96 

Jordan MS, Drury S, Giles JR, Manne S, Huang H, Chen Z, Oldridge D, Wherry EJ and Baxter AE: TET2 controls differentiation of terminally exhausted CD8 T cells. J Immunol. 206(1 Suppl): S14.072021. View Article : Google Scholar

97 

Liu B, Hu X, Feng K, Gao R, Xue Z, Zhang S, Zhang Y, Corse E, Hu Y, Han W and Zhang Z: Temporal single-cell tracing reveals clonal revival and expansion of precursor exhausted T cells during anti-PD-1 therapy in lung cancer. Nat Cancer. 3:108–121. 2022. View Article : Google Scholar

98 

Luoma AM, Suo S, Wang Y, Gunasti L, Porter CBM, Nabilsi N, Tadros J, Ferretti AP, Liao S, Gurer C, et al: Tissue-resident memory and circulating T cells are early responders to pre-surgical cancer immunotherapy. Cell. 185:2918–2935.e29. 2022. View Article : Google Scholar : PubMed/NCBI

99 

Li Z, Tuong ZK, Dean I, Willis C, Gaspal F, Fiancette R, Idris S, Kennedy B, Ferdinand JR, Peñalver A, et al: In vivo labeling reveals continuous trafficking of TCF-1+ T cells between tumor and lymphoid tissue. J Exp Med. 219:e202107492022. View Article : Google Scholar : PubMed/NCBI

100 

Kennedy BC, Dean I and Withers DR: Migration of stem-like CD8 T cells between tissue microenvironments underpins successful anti-tumour immune responses. Discov Immunol. 2:kyad0042023. View Article : Google Scholar : PubMed/NCBI

101 

Fransen MF, Schoonderwoerd M, Knopf P, Camps MG, Hawinkels LJ, Kneilling M, van Hall T and Ossendorp F: Tumor-draining lymph nodes are pivotal in PD-1/PD-L1 checkpoint therapy. JCI Insight. 3:e1245072018. View Article : Google Scholar : PubMed/NCBI

102 

Liu Z, Yu Z, Chen D, Verma V, Yuan C, Wang M, Wang F, Fan Q, Wang X, Li Y, et al: Pivotal roles of tumor-draining lymph nodes in the abscopal effects from combined immunotherapy and radiotherapy. Cancer Commun (Lond). 42:971–986. 2022. View Article : Google Scholar : PubMed/NCBI

103 

Tanaka R, Hiramitsu M, Shimizu S, Kawashima S, Sato A and Iwase Y: Efficient drug delivery to lymph nodes by intradermal administration and enhancement of anti-tumor effects of immune checkpoint inhibitors. Cancer Treat Res Commun. 36:1007402023. View Article : Google Scholar : PubMed/NCBI

104 

du Bois H, Heim TA and Lund AW: Tumor-draining lymph nodes: At the crossroads of metastasis and immunity. Sci Immunol. 6:eabg35512021. View Article : Google Scholar : PubMed/NCBI

105 

Rahim MK, Okholm TLH, Jones KB, McCarthy EE, Liu CC, Yee JL, Ki SJ, Marquez DM, Tenvooren I, Wai K, et al: Dynamic CD8+ T cell responses to cancer immunotherapy in human regional lymph nodes are disrupted in metastatic lymph nodes. Cell. 186:1127–1143.e18. 2023. View Article : Google Scholar

106 

Buchwald ZS, Nasti TH, Lee J, Eberhardt CS, Wieland A, Im SJ, Lawson D, Curran W, Ahmed R and Khan MK: Tumor-draining lymph node is important for a robust abscopal effect stimulated by radiotherapy. J Immunother Cancer. 8:e0008672020. View Article : Google Scholar : PubMed/NCBI

107 

Fear VS, Forbes CA, Neeve SA, Fisher SA, Chee J, Waithman J, Ma SK, Lake R, Nowak AK, Creaney J, et al: Tumour draining lymph node-generated CD8 T cells play a role in controlling lung metastases after a primary tumour is removed but not when adjuvant immunotherapy is used. Cancer Immunol Immunother. 70:3249–3258. 2021. View Article : Google Scholar : PubMed/NCBI

108 

Dammeijer F, van Gulijk M, Mulder EE, Lukkes M, Klaase L, van den Bosch T, van Nimwegen M, Lau SP, Latupeirissa K, Schetters S, et al: The PD-1/PD-L1-checkpoint restrains T cell immunity in tumor-draining lymph nodes. Cancer Cell. 38:685–700.e8. 2020. View Article : Google Scholar

109 

Zhou Y, Slone N, Chrisikos TT, Kyrysyuk O, Babcock RL, Medik YB, Li HS, Kleinerman ES and Watowich SS: Vaccine efficacy against primary and metastatic cancer with in vitro-generated CD103+ conventional dendritic cells. J Immunother Cancer. 8:e0004742020. View Article : Google Scholar

110 

Salmon H, Idoyaga J, Rahman A, Leboeuf M, Remark R, Jordan S, Casanova-Acebes M, Khudoynazarova M, Agudo J, Tung N, et al: Expansion and activation of CD103(+) dendritic cell progenitors at the tumor site enhances tumor responses to therapeutic PD-L1 and BRAF inhibition. Immunity. 44:924–938. 2016. View Article : Google Scholar : PubMed/NCBI

111 

Huang Q, Wu X, Wang Z, Chen X, Wang L, Lu Y, Xiong D, Liu Q, Tian Y, Lin H, 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. 185:4049–4066.e25. 2022. View Article : Google Scholar

112 

Okamura K, Nagayama S, Tate T, Chan HT, Kiyotani K and Nakamura Y: Lymphocytes in tumor-draining lymph nodes co-cultured with autologous tumor cells for adoptive cell therapy. J Transl Med. 20:2412022. View Article : Google Scholar : PubMed/NCBI

113 

Schenkel JM, Herbst RH, Canner D, Li A, Hillman M, Shanahan SL, Gibbons G, Smith OC, Kim JY, Westcott P, et al: Conventional type I dendritic cells maintain a reservoir of proliferative tumor-antigen specific TCF-1+ CD8+ T cells in tumor-draining lymph nodes. Immunity. 54:2338–2353.e6. 2021. View Article : Google Scholar

114 

Connolly KA, Kuchroo M, Venkat A, Khatun A, Wang J, William I, Hornick NI, Fitzgerald BL, Damo M, Kasmani MY, et al: A reservoir of stem-like CD8+ T cells in the tumor-draining lymph node preserves the ongoing antitumor immune response. Sci Immunol. 6:eabg78362021. View Article : Google Scholar

115 

O'Melia MJ, Manspeaker MP and Thomas SN: Tumor-draining lymph nodes are survival niches that support T cell priming against lymphatic transported tumor antigen and effects of immune checkpoint blockade in TNBC. Cancer Immunol Immunother. 70:2179–2195. 2021. View Article : Google Scholar : PubMed/NCBI

116 

Dominguez-Gutierrez PR, Kwenda EP, Donelan W, Miranda M, Doty A, O'Malley P, Crispen PL and Kusmartsev S: Detection of PD-L1-expressing myeloid cell clusters in the hyaluronan-enriched stroma in tumor tissue and tumor-draining lymph nodes. J Immunol. 208:2829–2836. 2022. View Article : Google Scholar : PubMed/NCBI

117 

Núñez NG, Tosello Boari J, Ramos RN, Richer W, Cagnard N, Anderfuhren CD, Niborski LL, Bigot J, Meseure D, De La Rochere P, et al: Tumor invasion in draining lymph nodes is associated with Treg accumulation in breast cancer patients. Nat Commun. 11:32722020. View Article : Google Scholar : PubMed/NCBI

118 

Yang H, Sun B, Ma W, Fan L, Xu K, Jia Y, Xu J, Wang Z and Yao F: Multi-scale characterization of tumor-draining lymph nodes in resectable lung cancer treated with neoadjuvant immune checkpoint inhibitors. EBioMedicine. 84:1042652022. View Article : Google Scholar : PubMed/NCBI

119 

Schumacher TN and Thommen DS: Tertiary lymphoid structures in cancer. Science. 375:eabf94192022. View Article : Google Scholar : PubMed/NCBI

120 

Dieu-Nosjean MC, Giraldo NA, Kaplon H, Germain C, Fridman WH and Sautès-Fridman C: Tertiary lymphoid structures, drivers of the anti-tumor responses in human cancers. Immunol Rev. 271:260–275. 2016. View Article : Google Scholar : PubMed/NCBI

121 

Rodriguez AB, Peske JD, Woods AN, Leick KM, Mauldin IS, Meneveau MO, Young SJ, Lindsay RS, Melssen MM, Cyranowski S, et al: Immune mechanisms orchestrate tertiary lymphoid structures in tumors via cancer-associated fibroblasts. Cell Rep. 36:1094222021. View Article : Google Scholar : PubMed/NCBI

122 

Ng KW, Boumelha J, Enfield KSS, Almagro J, Cha H, Pich O, Karasaki T, Moore DA, Salgado R, Sivakumar M, et al: Antibodies against endogenous retroviruses promote lung cancer immunotherapy. Nature. 616:563–573. 2023. View Article : Google Scholar : PubMed/NCBI

123 

Wang B, Liu J, Han Y, Deng Y, Li J and Jiang Y: The presence of tertiary lymphoid structures provides new insight into the clinicopathological features and prognosis of patients with breast cancer. Front Immunol. 13:8681552022. View Article : Google Scholar : PubMed/NCBI

124 

Wang Q, Shen X, An R, Bai J, Dong J, Cai H, Zhu H, Zhong W, Chen W, Liu A and Du J: Peritumoral tertiary lymphoid structure and tumor stroma percentage predict the prognosis of patients with non-metastatic colorectal cancer. Front Immunol. 13:9620562022. View Article : Google Scholar : PubMed/NCBI

125 

Zhang WH, Wang WQ, Han X, Gao HL, Xu SS, Li S, Li TJ, Xu HX, Li H, Ye LY, et al: Infiltrating pattern and prognostic value of tertiary lymphoid structures in resected non-functional pancreatic neuroendocrine tumors. J Immunother Cancer. 8:e0011882020. View Article : Google Scholar : PubMed/NCBI

126 

Tanaka T, Masuda A, Inoue J, Hamada T, Ikegawa T, Toyama H, Sofue K, Shiomi H, Sakai A, Kobayashi T, et al: Integrated analysis of tertiary lymphoid structures in relation to tumor-infiltrating lymphocytes and patient survival in pancreatic ductal adenocarcinoma. J Gastroenterol. 58:277–291. 2023. View Article : Google Scholar : PubMed/NCBI

127 

Ukita M, Hamanishi J, Yoshitomi H, Yamanoi K, Takamatsu S, Ueda A, Suzuki H, Hosoe Y, Furutake Y, Taki M, et al: CXCL13-producing CD4+ T cells accumulate in the early phase of tertiary lymphoid structures in ovarian cancer. JCI Insight. 7:e1572152022. View Article : Google Scholar : PubMed/NCBI

128 

Siliņa K, Soltermann A, Attar FM, Casanova R, Uckeley ZM, Thut H, Wandres M, Isajevs S, Cheng P, Curioni-Fontecedro A, et al: Germinal centers determine the prognostic relevance of tertiary lymphoid structures and are impaired by corticosteroids in lung squamous cell carcinoma. Cancer Res. 78:1308–1320. 2018. View Article : Google Scholar

129 

Yang M, Lu J, Zhang G, Wang Y, He M, Xu Q, Xu C and Liu H: CXCL13 shapes immunoactive tumor microenvironment and enhances the efficacy of PD-1 checkpoint blockade in high-grade serous ovarian cancer. J Immunother Cancer. 9:e0011362021. View Article : Google Scholar : PubMed/NCBI

130 

Cabrita R, Lauss M, Sanna A, Donia M, Skaarup Larsen M, Mitra S, Johansson I, Phung B, Harbst K, Vallon-Christersson J, et al: Tertiary lymphoid structures improve immunotherapy and survival in melanoma. Nature. 577:561–565. 2020. View Article : Google Scholar : PubMed/NCBI

131 

Helmink BA, Reddy SM, Gao J, Zhang S, Basar R, Thakur R, Yizhak K, Sade-Feldman M, Blando J, Han G, et al: B cells and tertiary lymphoid structures promote immunotherapy response. Nature. 577:549–555. 2020. View Article : Google Scholar : PubMed/NCBI

132 

Petitprez F, de Reyniès A, Keung EZ, Chen TW, Sun CM, Calderaro J, Jeng YM, Hsiao LP, Lacroix L, Bougoüin A, et al: B cells are associated with survival and immunotherapy response in sarcoma. Nature. 577:556–560. 2020. View Article : Google Scholar : PubMed/NCBI

133 

Sawada J, Hiraoka N, Qi R, Jiang L, Fournier-Goss AE, Yoshida M, Kawashima H and Komatsu M: Molecular signature of tumor-associated high endothelial venules that can predict breast cancer survival. Cancer Immunol Res. 10:468–481. 2022. View Article : Google Scholar : PubMed/NCBI

134 

Martinet L, Garrido I, Filleron T, Le Guellec S, Bellard E, Fournie JJ, Rochaix P and Girard JP: Human solid tumors contain high endothelial venules: association with T- and B-lymphocyte infiltration and favorable prognosis in breast cancer. Cancer Res. 71:5678–5687. 2011. View Article : Google Scholar : PubMed/NCBI

135 

Asrir A, Tardiveau C, Coudert J, Laffont R, Blanchard L, Bellard E, Veerman K, Bettini S, Lafouresse F, Vina E, et al: Tumor-associated high endothelial venules mediate lymphocyte entry into tumors and predict response to PD-1 plus CTLA-4 combination immunotherapy. Cancer Cell. 40:318–334.e9. 2022. View Article : Google Scholar : PubMed/NCBI

136 

Li Z, Jiang Y, Li B, Han Z, Shen J, Xia Y and Li R: Development and validation of a machine learning model for detection and classification of tertiary lymphoid structures in gastrointestinal cancers. JAMA Netw Open. 6:e22525532023. View Article : Google Scholar : PubMed/NCBI

137 

Neyt K, Perros F, GeurtsvanKessel CH, Hammad H and Lambrecht BN: Tertiary lymphoid organs in infection and autoimmunity. Trends Immunol. 33:297–305. 2012. View Article : Google Scholar : PubMed/NCBI

138 

Hayashi Y, Makino T, Sato E, Ohshima K, Nogi Y, Kanemura T, Honma K, Yamashita K, Saito T, Tanaka K, et al: Density and maturity of peritumoral tertiary lymphoid structures in oesophageal squamous cell carcinoma predicts patient survival and response to immune checkpoint inhibitors. Br J Cancer. 128:2175–2185. 2023. View Article : Google Scholar : PubMed/NCBI

139 

Deguchi S, Tanaka H, Suzuki S, Natsuki S, Mori T, Miki Y, Yoshii M, Tamura T, Toyokawa T, Lee S, et al: Clinical relevance of tertiary lymphoid structures in esophageal squamous cell carcinoma. BMC Cancer. 22:6992022. View Article : Google Scholar : PubMed/NCBI

140 

Ling Y, Zhong J, Weng Z, Lin G, Liu C, Pan C, Yang H, Wei X, Xie X, Wei X, et al: The prognostic value and molecular properties of tertiary lymphoid structures in oesophageal squamous cell carcinoma. Clin Transl Med. 12:e10742022. View Article : Google Scholar : PubMed/NCBI

141 

Calderaro J, Petitprez F, Becht E, Laurent A, Hirsch TZ, Rousseau B, Luciani A, Amaddeo G, Derman J, Charpy C, et al: Intra-tumoral tertiary lymphoid structures are associated with a low risk of early recurrence of hepatocellular carcinoma. J Hepatol. 70:58–65. 2019. View Article : Google Scholar

142 

Sun X, Liu W, Sun L, Mo H, Feng Y, Wu X, Li C, Chen C, Li J, Xin Y, et al: Maturation and abundance of tertiary lymphoid structures are associated with the efficacy of neoadjuvant chemoimmunotherapy in resectable non-small cell lung cancer. J Immunother Cancer. 10:e0055312022. View Article : Google Scholar :

143 

Lynch KT, Young SJ, Meneveau MO, Wages NA, Engelhard VH, Slingluff CL Jr and Mauldin IS: Heterogeneity in tertiary lymphoid structure B-cells correlates with patient survival in metastatic melanoma. J Immunother Cancer. 9:e0022732021. View Article : Google Scholar : PubMed/NCBI

144 

Posch F, Silina K, Leibl S, Mündlein A, Moch H, Siebenhüner A, Samaras P, Riedl J, Stotz M, Szkandera J, et al: Maturation of tertiary lymphoid structures and recurrence of stage II and III colorectal cancer. Oncoimmunology. 7:e13788442017. View Article : Google Scholar

145 

Zhang Q and Wu S: Tertiary lymphoid structures are critical for cancer prognosis and therapeutic response. Front Immunol. 13:10637112023. View Article : Google Scholar : PubMed/NCBI

146 

Fridman WH, Meylan M, Petitprez F, Sun CM, Italiano A and Sautès-Fridman C: B cells and tertiary lymphoid structures as determinants of tumour immune contexture and clinical outcome. Nat Rev Clin Oncol. 19:441–457. 2022. View Article : Google Scholar : PubMed/NCBI

147 

Saito T, Nishikawa H, Wada H, Nagano Y, Sugiyama D, Atarashi K, Maeda Y, Hamaguchi M, Ohkura N, Sato E, et al: Two FOXP3(+)CD4(+) T cell subpopulations distinctly control the prognosis of colorectal cancers. Nat Med. 22:679–684. 2016. View Article : Google Scholar : PubMed/NCBI

148 

Shalapour S, Font-Burgada J, Di Caro G, Zhong Z, Sanchez-Lopez E, Dhar D, Willimsky G, Ammirante M, Strasner A, Hansel DE, et al: Immunosuppressive plasma cells impede T-cell-dependent immunogenic chemotherapy. Nature. 521:94–98. 2015. View Article : Google Scholar : PubMed/NCBI

149 

Finkin S, Yuan D, Stein I, Taniguchi K, Weber A, Unger K, Browning JL, Goossens N, Nakagawa S, Gunasekaran G, et al: Ectopic lymphoid structures function as microniches for tumor progenitor cells in hepatocellular carcinoma. Nat Immunol. 16:1235–1244. 2015. View Article : Google Scholar : PubMed/NCBI

150 

Milutinovic S, Abe J, Godkin A, Stein JV and Gallimore A: The dual role of high endothelial venules in cancer progression versus immunity. Trends Cancer. 7:214–225. 2021. View Article : Google Scholar

151 

Sautès-Fridman C, Petitprez F, Calderaro J and Fridman WH: Tertiary lymphoid structures in the era of cancer immunotherapy. Nat Rev Cancer. 19:307–325. 2019. View Article : Google Scholar : PubMed/NCBI

152 

Ray-Coquard I, Cropet C, Van Glabbeke M, Sebban C, Le Cesne A, Judson I, Tredan O, Verweij J, Biron P, Labidi I, et al: Lymphopenia as a prognostic factor for overall survival in advanced carcinomas, sarcomas, and lymphomas. Cancer Res. 69:5383–5391. 2009. View Article : Google Scholar : PubMed/NCBI

153 

Wu Z, Zhang J, Cai Y, Deng R, Yang L, Li J and Deng Y: Reduction of circulating lymphocyte count is a predictor of good tumor response after neoadjuvant treatment for rectal cancer. Medicine (Baltimore). 97:e114352018. View Article : Google Scholar : PubMed/NCBI

154 

Lee YJ, Park YS, Lee HW, Park TY, Lee JK and Heo EY: Peripheral lymphocyte count as a surrogate marker of immune checkpoint inhibitor therapy outcomes in patients with non-small-cell lung cancer. Sci Rep. 12:6262022. View Article : Google Scholar : PubMed/NCBI

155 

Weide B, Martens A, Hassel JC, Berking C, Postow MA, Bisschop K, Simeone E, Mangana J, Schilling B, Di Giacomo AM, et al: Baseline biomarkers for outcome of melanoma patients treated with pembrolizumab. Clin Cancer Res. 22:5487–5496. 2016. View Article : Google Scholar : PubMed/NCBI

156 

Martens A, Wistuba-Hamprecht K, Geukes Foppen M, Yuan J, Postow MA, Wong P, Romano E, Khammari A, Dreno B, Capone M, et al: Baseline peripheral blood biomarkers associated with clinical outcome of advanced melanoma patients treated with ipilimumab. Clin Cancer Res. 22:2908–2918. 2016. View Article : Google Scholar : PubMed/NCBI

157 

Juliá EP, Mandó P, Rizzo MM, Cueto GR, Tsou F, Luca R, Pupareli C, Bravo AI, Astorino W, Mordoh J, et al: Peripheral changes in immune cell populations and soluble mediators after anti-PD-1 therapy in non-small cell lung cancer and renal cell carcinoma patients. Cancer Immunol Immunother. 68:1585–1596. 2019. View Article : Google Scholar : PubMed/NCBI

158 

Cui JH, Lin KR, Yuan SH, Jin YB, Chen XP, Su XK, Jiang J, Pan YM, Mao SL, Mao XF and Luo W: TCR repertoire as a novel indicator for immune monitoring and prognosis assessment of patients with cervical cancer. Front Immunol. 9:27292018. View Article : Google Scholar : PubMed/NCBI

159 

Gleason L, Porcu P and Nikbakht N: Reduced overall T-cell receptor diversity as an indicator of aggressive cutaneous T-cell lymphoma. Blood. 140(Suppl 1): 3539–3540. 2022. View Article : Google Scholar

160 

Manuel M, Tredan O, Bachelot T, Clapisson G, Courtier A, Parmentier G, Rabeony T, Grives A, Perez S, Mouret JF, et al: Lymphopenia combined with low TCR diversity (divpenia) predicts poor overall survival in metastatic breast cancer patients. Oncoimmunology. 1:432–440. 2012. View Article : Google Scholar : PubMed/NCBI

161 

Lin KR, Pang DM, Jin YB, Hu Q, Pan YM, Cui JH, Chen XP, Lin YX, Mao XF, Duan HB and Luo W: Circulating CD8+ T-cell repertoires reveal the biological characteristics of tumors and clinical responses to chemotherapy in breast cancer patients. Cancer Immunol Immunother. 67:1743–1752. 2018. View Article : Google Scholar : PubMed/NCBI

162 

Cai G, Guan Z, Jin Y, Su Z, Chen X, Liu Q, Wang C, Yin X, Zhang L, Ye G and Luo W: Circulating T-cell repertoires correlate with the tumor response in patients with breast cancer receiving neoadjuvant chemotherapy. JCO Precis Oncol. 6:e21001202022. View Article : Google Scholar : PubMed/NCBI

163 

Page DB, Yuan J, Redmond D, Wen YH, Durack JC, Emerson R, Solomon S, Dong Z, Wong P, Comstock C, et al: Deep sequencing of T-cell receptor DNA as a biomarker of clonally expanded TILs in breast cancer after immunotherapy. Cancer Immunol Res. 4:835–844. 2016. View Article : Google Scholar : PubMed/NCBI

164 

Kuehm LM, Wolf K, Zahour J, DiPaolo RJ and Teague RM: Checkpoint blockade immunotherapy enhances the frequency and effector function of murine tumor-infiltrating T cells but does not alter TCRβ diversity. Cancer Immunol Immunother. 68:1095–1106. 2019. View Article : Google Scholar : PubMed/NCBI

165 

Rudqvist NP, Pilones KA, Lhuillier C, Wennerberg E, Sidhom JW, Emerson RO, Robins HS, Schneck J, Formenti SC and Demaria S: Radiotherapy and CTLA-4 blockade shape the TCR repertoire of tumor-infiltrating T cells. Cancer Immunol Res. 6:139–150. 2018. View Article : Google Scholar :

166 

Wu TD, Madireddi S, de Almeida PE, Banchereau R, Chen YJ, Chitre AS, Chiang EY, Iftikhar H, O'Gorman WE, Au-Yeung A, et al: Peripheral T cell expansion predicts tumour infiltration and clinical response. Nature. 579:274–278. 2020. View Article : Google Scholar : PubMed/NCBI

167 

Puig-Saus C, Sennino B, Peng S, Wang CL, Pan Z, Yuen B, Purandare B, An D, Quach BB, Nguyen D, et al: Neoantigen-targeted CD8+ T cell responses with PD-1 blockade therapy. Nature. 615:697–704. 2023. View Article : Google Scholar : PubMed/NCBI

168 

Dong N, Moreno-Manuel A, Calabuig-Fariñas S, Gallach S, Zhang F, Blasco A, Aparisi F, Meri-Abad M, Guijarro R, Sirera R, et al: Characterization of circulating T cell receptor repertoire provides information about clinical outcome after PD-1 blockade in advanced non-small cell lung cancer patients. Cancers (Basel). 13:29502021. View Article : Google Scholar : PubMed/NCBI

169 

Han J, Duan J, Bai H, Wang Y, Wan R, Wang X, Chen S, Tian Y, Wang D, Fei K, et al: TCR repertoire diversity of peripheral PD-1+CD8+ T cells predicts clinical outcomes after immunotherapy in patients with non-small lung cancer. Cancer Immunol Res. 8:146–154. 2020. View Article : Google Scholar

170 

Kato T, Kiyotani K, Tomiyama E, Koh Y, Matsushita M, Hayashi Y, Nakano K, Ishizuya Y, Wang C, Hatano K, et al: Peripheral T cell receptor repertoire features predict durable responses to anti-PD-1 inhibitor monotherapy in advanced renal cell carcinoma. Oncoimmunology. 10:18629482021. View Article : Google Scholar : PubMed/NCBI

171 

Snyder A, Nathanson T, Funt SA, Ahuja A, Buros Novik J, Hellmann MD, Chang E, Aksoy BA, Al-Ahmadie H, Yusko E, et al: Contribution of systemic and somatic factors to clinical response and resistance to PD-L1 blockade in urothelial cancer: An exploratory multi-omic analysis. PLoS Med. 14:e10023092017. View Article : Google Scholar : PubMed/NCBI

172 

Arakawa A, Vollmer S, Tietze J, Galinski A, Heppt MV, Bürdek M, Berking C and Prinz JC: Clonality of CD4+ blood T cells predicts longer survival with CTLA4 or PD-1 checkpoint inhibition in advanced melanoma. Front Immunol. 10:13362019. View Article : Google Scholar

173 

Zhu Q, Qiao G, Huang L, Xu C, Guo D, Wang S, Zhao J, Song Y, Liu B, Chen Z, et al: Restored CD8+PD-1+ T cells facilitate the response to Anti-PD-1 for patients with pancreatic ductal adenocarcinoma. Front Oncol. 12:8375602022. View Article : Google Scholar

174 

Verronèse E, Delgado A, Valladeau-Guilemond J, Garin G, Guillemaut S, Tredan O, Ray-Coquard I, Bachelot T, N'Kodia A, Bardin-Dit-Courageot C, et al: Immune cell dysfunctions in breast cancer patients detected through whole blood multi-parametric flow cytometry assay. Oncoimmunology. 5:e11007912015. View Article : Google Scholar

175 

Saleh R and Elkord E: FoxP3+ T regulatory cells in cancer: Prognostic biomarkers and therapeutic targets. Cancer Lett. 490:174–185. 2020. View Article : Google Scholar : PubMed/NCBI

176 

Sakaguchi S, Miyara M, Costantino CM and Hafler DA: FOXP3+ regulatory T cells in the human immune system. Nat Rev Immunol. 10:490–500. 2010. View Article : Google Scholar : PubMed/NCBI

177 

Kotsakis A, Koinis F, Katsarou A, Gioulbasani M, Aggouraki D, Kentepozidis N, Georgoulias V and Vetsika EK: Prognostic value of circulating regulatory T cell subsets in untreated non-small cell lung cancer patients. Sci Rep. 6:392472016. View Article : Google Scholar : PubMed/NCBI

178 

Mamessier E, Sylvain A, Thibult ML, Houvenaeghel G, Jacquemier J, Castellano R, Gonçalves A, André P, Romagné F, Thibault G, et al: Human breast cancer cells enhance self tolerance by promoting evasion from NK cell antitumor immunity. J Clin Invest. 121:3609–3622. 2011. View Article : Google Scholar : PubMed/NCBI

179 

Manjarrez-Orduño N, Menard LC, Kansal S, Fischer P, Kakrecha B, Jiang C, Cunningham M, Greenawalt D, Patel V, Yang M, et al: Circulating T cell subpopulations correlate with immune responses at the tumor site and clinical response to PD1 inhibition in non-small cell lung cancer. Front Immunol. 9:16132018. View Article : Google Scholar : PubMed/NCBI

180 

Kim CG, Kim KH, Pyo KH, Xin CF, Hong MH, Ahn BC, Kim Y, Choi SJ, Yoon HI, Lee JG, et al: Hyperprogressive disease during PD-1/PD-L1 blockade in patients with non-small-cell lung cancer. Ann Oncol. 30:1104–1113. 2019. View Article : Google Scholar : PubMed/NCBI

181 

Wistuba-Hamprecht K, Martens A, Heubach F, Romano E, Geukes Foppen M, Yuan J, Postow M, Wong P, Mallardo D, Schilling B, et al: Peripheral CD8 effector-memory type 1 T-cells correlate with outcome in ipilimumab-treated stage IV melanoma patients. Eur J Cancer. 73:61–70. 2017. View Article : Google Scholar : PubMed/NCBI

182 

Kagamu H, Kitano S, Yamaguchi O, Yoshimura K, Horimoto K, Kitazawa M, Fukui K, Shiono A, Mouri A, Nishihara F, et al: CD4+ T-cell Immunity in the peripheral blood correlates with response to Anti-PD-1 therapy. Cancer Immunol Res. 8:334–344. 2020. View Article : Google Scholar

183 

Sade-Feldman M, Kanterman J, Klieger Y, Ish-Shalom E, Olga M, Saragovi A, Shtainberg H, Lotem M and Baniyash M: Clinical significance of circulating CD33+CD11b+HLA-DR-myeloid cells in patients with stage IV melanoma treated with ipilimumab. Clin Cancer Res. 22:5661–5672. 2016. View Article : Google Scholar : PubMed/NCBI

184 

Zuazo M, Arasanz H, Fernández-Hinojal G, García-Granda MJ, Gato M, Bocanegra A, Martínez M, Hernández B, Teijeira L, Morilla I, et al: Functional systemic CD4 immunity is required for clinical responses to PD-L1/PD-1 blockade therapy. EMBO Mol Med. 11:e102932019. View Article : Google Scholar : PubMed/NCBI

185 

Mazzaschi G, Facchinetti F, Missale G, Canetti D, Madeddu D, Zecca A, Veneziani M, Gelsomino F, Goldoni M, Buti S, et al: The circulating pool of functionally competent NK and CD8+ cells predicts the outcome of anti-PD1 treatment in advanced NSCLC. Lung Cancer. 127:153–163. 2019. View Article : Google Scholar : PubMed/NCBI

186 

Kamada T, Togashi Y, Tay C, Ha D, Sasaki A, Nakamura Y, Sato E, Fukuoka S, Tada Y, Tanaka A, et al: PD-1+ regulatory T cells amplified by PD-1 blockade promote hyperprogression of cancer. Proc Natl Acad Sci USA. 116:9999–10008. 2019. View Article : Google Scholar

187 

Jacquelot N, Roberti MP, Enot DP, Rusakiewicz S, Ternès N, Jegou S, Woods DM, Sodré AL, Hansen M, Meirow Y, et al: Predictors of responses to immune checkpoint blockade in advanced melanoma. Nat Commun. 8:5922017. View Article : Google Scholar : PubMed/NCBI

188 

Ferrara R, Naigeon M, Auclin E, Duchemann B, Cassard L, Jouniaux JM, Boselli L, Grivel J, Desnoyer A, Mezquita L, et al: Circulating T-cell immunosenescence in patients with advanced non-small cell lung cancer treated with single-agent PD-1/PD-L1 inhibitors or platinum-based chemotherapy. Clin Cancer Res. 27:492–503. 2021. View Article : Google Scholar

189 

Griffiths JI, Wallet P, Pflieger LT, Stenehjem D, Liu X, Cosgrove PA, Leggett NA, McQuerry JA, Shrestha G, Rossetti M, et al: Circulating immune cell phenotype dynamics reflect the strength of tumor-immune cell interactions in patients during immunotherapy. Proc Natl Acad Sci USA. 117:16072–16082. 2020. View Article : Google Scholar : PubMed/NCBI

190 

Huang AC, Postow MA, Orlowski RJ, Mick R, Bengsch B, Manne S, Xu W, Harmon S, Giles JR, Wenz B, et al: T-cell invigoration to tumour burden ratio associated with anti-PD-1 response. Nature. 545:60–65. 2017. View Article : Google Scholar : PubMed/NCBI

191 

Kwon M, An M, Klempner SJ, Lee H, Kim KM, Sa JK, Cho HJ, Hong JY, Lee T, Min YW, et al: Determinants of response and intrinsic resistance to PD-1 blockade in microsatellite instability-high gastric cancer. Cancer Discov. 11:2168–2185. 2021. View Article : Google Scholar : PubMed/NCBI

192 

Kamphorst AO, Pillai RN, Yang S, Nasti TH, Akondy RS, Wieland A, Sica GL, Yu K, Koenig L, Patel NT, et al: Proliferation of PD-1+ CD8 T cells in peripheral blood after PD-1-targeted therapy in lung cancer patients. Proc Natl Acad Sci USA. 114:4993–4998. 2017. View Article : Google Scholar : PubMed/NCBI

193 

Kim KH, Cho J, Ku BM, Koh J, Sun JM, Lee SH, Ahn JS, Cheon J, Min YJ, Park SH, et al: The first-week proliferative response of peripheral blood PD-1+CD8+ T cells predicts the response to anti-PD-1 therapy in solid tumors. Clin Cancer Res. 25:2144–2154. 2019. View Article : Google Scholar : PubMed/NCBI

194 

Krieg C, Nowicka M, Guglietta S, Schindler S, Hartmann FJ, Weber LM, Dummer R, Robinson MD, Levesque MP and Becher B: High-dimensional single-cell analysis predicts response to anti-PD-1 immunotherapy. Nat Med. 24:144–153. 2018. View Article : Google Scholar : PubMed/NCBI

195 

de Coaña YP, Wolodarski M, Poschke I, Yoshimoto Y, Yang Y, Nyström M, Edbäck U, Brage SE, Lundqvist A, Masucci GV, et al: Ipilimumab treatment decreases monocytic MDSCs and increases CD8 effector memory T cells in long-term survivors with advanced melanoma. Oncotarget. 8:21539–21553. 2017. View Article : Google Scholar : PubMed/NCBI

196 

Duchemann B, Naigeon M, Auclin E, Ferrara R, Cassard L, Jouniaux JM, Boselli L, Grivel J, Desnoyer A, Danlos FX, et al: CD8+PD-1+ to CD4+PD-1+ ratio (PERLS) is associated with prognosis of patients with advanced NSCLC treated with PD-(L)1 blockers. J Immunother Cancer. 10:e0040122022. View Article : Google Scholar

197 

Youn JI, Park SM, Park S, Kim G, Lee HJ, Son J, Hong MH, Ghaderpour A, Baik B, Islam J, et al: Peripheral natural killer cells and myeloid-derived suppressor cells correlate with anti-PD-1 responses in non-small cell lung cancer. Sci Rep. 10:90502020. View Article : Google Scholar : PubMed/NCBI

198 

Dodagatta-Marri E, Meyer DS, Reeves MQ, Paniagua R, To MD, Binnewies M, Broz ML, Mori H, Wu D, Adoumie M, et al: α-PD-1 therapy elevates Treg/Th balance and increases tumor cell pSmad3 that are both targeted by α-TGFβ antibody to promote durable rejection and immunity in squamous cell carcinomas. J Immunother Cancer. 7:622019. View Article : Google Scholar

199 

Takeuchi Y, Tanemura A, Tada Y, Katayama I, Kumanogoh A and Nishikawa H: Clinical response to PD-1 blockade correlates with a sub-fraction of peripheral central memory CD4+ T cells in patients with malignant melanoma. Int Immunol. 30:13–22. 2018. View Article : Google Scholar : PubMed/NCBI

200 

Kato R, Yamasaki M, Urakawa S, Nishida K, Makino T, Morimoto-Okazawa A, Kawashima A, Iwahori K, Suzuki S, Ueda R, et al: Increased Tim-3+ T cells in PBMCs during nivolumab therapy correlate with responses and prognosis of advanced esophageal squamous cell carcinoma patients. Cancer Immunol Immunother. 67:1673–1683. 2018. View Article : Google Scholar : PubMed/NCBI

201 

Sangro B, Melero I, Wadhawan S, Finn RS, Abou-Alfa GK, Cheng AL, Yau T, Furuse J, Park JW, Boyd Z, et al: Association of inflammatory biomarkers with clinical outcomes in nivolumab-treated patients with advanced hepatocellular carcinoma. J Hepatol. 73:1460–1469. 2020. View Article : Google Scholar : PubMed/NCBI

202 

Kim HR, Park SM, Seo SU, Jung I, Yoon HI, Gabrilovich DI, Cho BC, Seong SY, Ha SJ and Youn JI: The ratio of peripheral regulatory T cells to Lox-1+ polymorphonuclear myeloid-derived suppressor cells predicts the early response to Anti-PD-1 therapy in patients with non-small cell lung cancer. Am J Respir Crit Care Med. 199:243–246. 2019. View Article : Google Scholar :

203 

Jia XH, Geng LY, Jiang PP, Xu H, Nan KJ, Yao Y, Jiang LL, Sun H, Qin TJ and Guo H: The biomarkers related to immune related adverse events caused by immune checkpoint inhibitors. J Exp Clin Cancer Res. 39:2842020. View Article : Google Scholar : PubMed/NCBI

204 

Schweizer C, Schubert P, Rutzner S, Eckstein M, Haderlein M, Lettmaier S, Semrau S, Gostian AO, Frey B, Gaipl US, et al: Prospective evaluation of the prognostic value of immune-related adverse events in patients with non-melanoma solid tumour treated with PD-1/PD-L1 inhibitors alone and in combination with radiotherapy. Eur J Cancer. 140:55–62. 2020. View Article : Google Scholar : PubMed/NCBI

205 

Chuah S, Lee J, Song Y, Kim HD, Wasser M, Kaya NA, Bang K, Lee YJ, Jeon SH, Suthen S, et al: Uncoupling immune trajectories of response and adverse events from anti-PD-1 immunotherapy in hepatocellular carcinoma. J Hepatol. 77:683–694. 2022. View Article : Google Scholar : PubMed/NCBI

206 

Zhu H, Galdos FX, Lee D, Waliany S, Huang YV, Ryan J, Dang K, Neal JW, Wakelee HA, Reddy SA, et al: Identification of pathogenic immune cell subsets associated with checkpoint inhibitor-induced myocarditis. Circulation. 146:316–335. 2022. View Article : Google Scholar : PubMed/NCBI

207 

Franken A, Van Mol P, Vanmassenhove S, Donders E, Schepers R, Van Brussel T, Dooms C, Yserbyt J, De Crem N, Testelmans D, et al: Single-cell transcriptomics identifies pathogenic T-helper 17.1 cells and pro-inflammatory monocytes in immune checkpoint inhibitor-related pneumonitis. J Immunother Cancer. 10:e0053232022. View Article : Google Scholar : PubMed/NCBI

208 

Kim KH, Hur JY, Cho J, Ku BM, Koh J, Koh JY, Sun JM, Lee SH, Ahn JS, Park K, et al: Immune-related adverse events are clustered into distinct subtypes by T-cell profiling before and early after anti-PD-1 treatment. Oncoimmunology. 9:17220232020. View Article : Google Scholar : PubMed/NCBI

209 

Wei SC, Meijers WC, Axelrod ML, Anang NAAS, Screever EM, Wescott EC, Johnson DB, Whitley E, Lehmann L, Courand PY, et al: A genetic mouse model recapitulates immune checkpoint inhibitor-associated myocarditis and supports a mechanism-based therapeutic intervention. Cancer Discov. 11:614–625. 2021. View Article : Google Scholar :

210 

Fairfax BP, Taylor CA, Watson RA, Nassiri I, Danielli S, Fang H, Mahé EA, Cooper R, Woodcock V, Traill Z, et al: Peripheral CD8+ T cell characteristics associated with durable responses to immune checkpoint blockade in patients with metastatic melanoma. Nat Med. 26:193–199. 2020. View Article : Google Scholar : PubMed/NCBI

211 

Chia S, Bedard PL, Hilton J, Amir E, Gelmon K, Goodwin R, Villa D, Cabanero M, Tu D, Tsao M and Seymour L: A phase Ib trial of durvalumab in combination with trastuzumab in HER2-positive metastatic breast cancer (CCTG IND.229). Oncologist. 24:1439–1445. 2019. View Article : Google Scholar : PubMed/NCBI

212 

Soler MF, Abaurrea A, Azcoaga P, Araujo AM and Caffarel MM: New perspectives in cancer immunotherapy: Targeting IL-6 cytokine family. J Immunother Cancer. 11:e0075302023. View Article : Google Scholar : PubMed/NCBI

213 

Wattenberg MM and Beatty GL: Overcoming immunotherapeutic resistance by targeting the cancer inflammation cycle. Semin Cancer Biol. 65:38–50. 2020. View Article : Google Scholar : PubMed/NCBI

214 

Tobias J, Steinberger P, Drinić M and Wiedermann U: Emerging targets for anticancer vaccination: PD-1. ESMO Open. 6:1002782021. View Article : Google Scholar : PubMed/NCBI

215 

Tang Q, Chen Y, Li X, Long S, Shi Y, Yu Y, Wu W, Han L and Wang S: The role of PD-1/PD-L1 and application of immune-checkpoint inhibitors in human cancers. Front Immunol. 13:9644422022. View Article : Google Scholar : PubMed/NCBI

216 

Yang Y, Yu Y and Lu S: Effectiveness of PD-1/PD-L1 inhibitors in the treatment of lung cancer: Brightness and challenge. Sci China Life Sci. 63:1499–1514. 2020. View Article : Google Scholar : PubMed/NCBI

217 

Reck M, Remon J and Hellmann MD: First-line immunotherapy for non-small-cell lung cancer. J Clin Oncol. 40:586–597. 2022. View Article : Google Scholar : PubMed/NCBI

218 

Schoenfeld AJ, Arbour KC, Rizvi H, Iqbal AN, Gadgeel SM, Girshman J, Kris MG, Riely GJ, Yu HA and Hellmann MD: Severe immune-related adverse events are common with sequential PD-(L)1 blockade and osimertinib. Ann Oncol. 30:839–844. 2019. View Article : Google Scholar : PubMed/NCBI

219 

Kennedy LB and Salama AKS: A review of cancer immunotherapy toxicity. CA Cancer J Clin. 70:86–104. 2020. View Article : Google Scholar : PubMed/NCBI

220 

Yi M, Zheng X, Niu M, Zhu S, Ge H and Wu K: Combination strategies with PD-1/PD-L1 blockade: Current advances and future directions. Mol Cancer. 21:282022. View Article : Google Scholar : PubMed/NCBI

221 

Boutros C, Tarhini A, Routier E, Lambotte O, Ladurie FL, Carbonnel F, Izzeddine H, Marabelle A, Champiat S, Berdelou A, et al: Safety profiles of anti-CTLA-4 and anti-PD-1 antibodies alone and in combination. Nat Rev Clin Oncol. 13:473–486. 2016. View Article : Google Scholar : PubMed/NCBI

222 

Jhawar SR, Wang SJ, Thandoni A, Bommareddy PK, Newman JH, Marzo AL, Kuzel TM, Gupta V, Reiser J, Daniels P, et al: Combination oncolytic virus, radiation therapy, and immune checkpoint inhibitor treatment in anti-PD-1-refractory cancer. J Immunother Cancer. 11:e0067802023. View Article : Google Scholar : PubMed/NCBI

223 

Gebrael G, Sahu KK, Agarwal N and Maughan BL: Update on combined immunotherapy for the treatment of advanced renal cell carcinoma. Hum Vaccin Immunother. 19:21935282023. View Article : Google Scholar : PubMed/NCBI

224 

Cafri G, Gartner JJ, Zaks T, Hopson K, Levin N, Paria BC, Parkhurst MR, Yossef R, Lowery FJ, Jafferji MS, et al: mRNA vaccine-induced neoantigen-specific T cell immunity in patients with gastrointestinal cancer. J Clin Invest. 130:5976–5988. 2020. View Article : Google Scholar : PubMed/NCBI

225 

Ott PA, Hu-Lieskovan S, Chmielowski B, Govindan R, Naing A, Bhardwaj N, Margolin K, Awad MM, Hellmann MD, Lin JJ, et al: A phase Ib trial of personalized neoantigen therapy plus anti-PD-1 in patients with advanced melanoma, non-small cell lung cancer, or bladder cancer. Cell. 183:347–362.e24. 2020. View Article : Google Scholar : PubMed/NCBI

226 

Keskin DB, Anandappa AJ, Sun J, Tirosh I, Mathewson ND, Li S, Oliveira G, Giobbie-Hurder A, Felt K, Gjini E, et al: Neoantigen vaccine generates intratumoral T cell responses in phase Ib glioblastoma trial. Nature. 565:234–239. 2019. View Article : Google Scholar :

227 

Platten M, Bunse L, Wick A, Bunse T, Le Cornet L, Harting I, Sahm F, Sanghvi K, Tan CL, Poschke I, et al: A vaccine targeting mutant IDH1 in newly diagnosed glioma. Nature. 592:463–468. 2021. View Article : Google Scholar : PubMed/NCBI

228 

Kochenderfer JN, Chien CD, Simpson JL and Gress RE: Maximizing CD8+ T cell responses elicited by peptide vaccines containing CpG oligodeoxynucleotides. Clin Immunol. 124:119–130. 2007. View Article : Google Scholar : PubMed/NCBI

229 

Haining WN, Davies J, Kanzler H, Drury L, Brenn T, Evans J, Angelosanto J, Rivoli S, Russell K, George S, et al: CpG oligodeoxynucleotides alter lymphocyte and dendritic cell trafficking in humans. Clin Cancer Res. 14:5626–5634. 2008. View Article : Google Scholar : PubMed/NCBI

230 

Song YC, Cheng HY, Leng CH, Chiang SK, Lin CW, Chong P, Huang MH and Liu SJ: A novel emulsion-type adjuvant containing CpG oligodeoxynucleotides enhances CD8+ T-cell-mediated anti-tumor immunity. J Control Release. 173:158–165. 2014. View Article : Google Scholar

231 

Baumgaertner P, Costa Nunes C, Cachot A, Maby-El Hajjami H, Cagnon L, Braun M, Derré L, Rivals JP, Rimoldi D, Gnjatic S, et al: Vaccination of stage III/IV melanoma patients with long NY-ESO-1 peptide and CpG-B elicits robust CD8+ and CD4+ T-cell responses with multiple specificities including a novel DR7-restricted epitope. Oncoimmunology. 5:e12162902016. View Article : Google Scholar

232 

Zhu P, Li SY, Ding J, Fei Z, Sun SN, Zheng ZH, Wei D, Jiang J, Miao JL, Li SZ, et al: Combination immunotherapy of glioblastoma with dendritic cell cancer vaccines, anti-PD-1 and poly I:C. J Pharm Anal. 13:616–624. 2023. View Article : Google Scholar : PubMed/NCBI

233 

Soltani M, Savvateeva LV, Ganjalikhani-Hakemi M and Zamyatnin AA: Clinical combinatorial treatments based on cancer vaccines: Combination with checkpoint inhibitors and beyond. Curr Drug Targets. 23:1072–1084. 2022. View Article : Google Scholar : PubMed/NCBI

234 

van der Burg SH, Arens R, Ossendorp F, van Hall T and Melief CJ: Vaccines for established cancer: Overcoming the challenges posed by immune evasion. Nat Rev Cancer. 16:219–233. 2016. View Article : Google Scholar : PubMed/NCBI

235 

Reichmuth AM, Oberli MA, Jaklenec A, Langer R and Blankschtein D: mRNA vaccine delivery using lipid nanoparticles. Ther Deliv. 7:319–334. 2016. View Article : Google Scholar : PubMed/NCBI

236 

Kon E, Ad-El N, Hazan-Halevy I, Stotsky-Oterin L and Peer D: Targeting cancer with mRNA-lipid nanoparticles: Key considerations and future prospects. Nat Rev Clin Oncol. 20:739–754. 2023. View Article : Google Scholar : PubMed/NCBI

237 

Tenchov R, Bird R, Curtze AE and Zhou Q: Lipid nanoparticles-from liposomes to mRNA vaccine delivery, a landscape of research diversity and advancement. ACS Nano. 15:16982–17015. 2021. View Article : Google Scholar : PubMed/NCBI

238 

Karikó K, Muramatsu H, Welsh FA, Ludwig J, Kato H, Akira S and Weissman D: Incorporation of pseudouridine into mRNA yields superior nonimmunogenic vector with increased translational capacity and biological stability. Mol Ther. 16:1833–1840. 2008. View Article : Google Scholar : PubMed/NCBI

239 

Ramos da Silva J, Bitencourt Rodrigues K, Formoso Pelegrin G, Silva Sales N, Muramatsu H, de Oliveira Silva M, Porchia BFMM, Moreno ACR, Aps LRMM, Venceslau-Carvalho AA, et al: Single immunizations of self-amplifying or non-replicating mRNA-LNP vaccines control HPV-associated tumors in mice. Sci Transl Med. 15:eabn34642023. View Article : Google Scholar : PubMed/NCBI

240 

Sittplangkoon C, Alameh MG, Weissman D, Lin PJC, Tam YK, Prompetchara E and Palaga T: mRNA vaccine with unmodified uridine induces robust type I interferon-dependent anti-tumor immunity in a melanoma model. Front Immunol. 13:9830002022. View Article : Google Scholar : PubMed/NCBI

241 

Meulewaeter S, Aernout I, Deprez J, Engelen Y, De Velder M, Franceschini L, Breckpot K, Van Calenbergh S, Asselman C, Boucher K, et al: Alpha-galactosylceramide improves the potency of mRNA LNP vaccines against cancer and intracellular bacteria. J Control Release. 370:379–391. 2024. View Article : Google Scholar : PubMed/NCBI

242 

Yang J, Arya S, Lung P, Lin Q, Huang J and Li Q: Hybrid nanovaccine for the co-delivery of the mRNA antigen and adjuvant. Nanoscale. 11:21782–21789. 2019. View Article : Google Scholar : PubMed/NCBI

243 

Carvalho T: Personalized anti-cancer vaccine combining mRNA and immunotherapy tested in melanoma trial. Nat Med. 29:2379–2380. 2023. View Article : Google Scholar : PubMed/NCBI

244 

Podaza E, Carri I, Aris M, von Euw E, Bravo AI, Blanco P, Ortiz Wilczyñski JM, Koile D, Yankilevich P, Nielsen M, et al: Evaluation of T-cell responses against shared melanoma associated antigens and predicted neoantigens in cutaneous melanoma patients treated with the CSF-470 allogeneic cell vaccine plus BCG and GM-CSF. Front Immunol. 11:11472020. View Article : Google Scholar : PubMed/NCBI

245 

Sebastian M, Schröder A, Scheel B, Hong HS, Muth A, von Boehmer L, Zippelius A, Mayer F, Reck M, Atanackovic D, et al: A phase I/IIa study of the mRNA-based cancer immunotherapy CV9201 in patients with stage IIIB/IV non-small cell lung cancer. Cancer Immunol Immunother. 68:799–812. 2019. View Article : Google Scholar : PubMed/NCBI

246 

Zilio S, Bicciato S, Weed D and Serafini P: CCR1 and CCR5 mediate cancer-induced myelopoiesis and differentiation of myeloid cells in the tumor. J Immunother Cancer. 10:e0031312022. View Article : Google Scholar : PubMed/NCBI

247 

Badr G, Al-Sadoon MK, Rabah DM and Sayed D: Snake (Walterinnesia aegyptia) venom-loaded silica nanoparticles induce apoptosis and growth arrest in human prostate cancer cells. Apoptosis. 18:300–314. 2013. View Article : Google Scholar

248 

Badr G, Al-Sadoon MK and Rabah DM: Therapeutic efficacy and molecular mechanisms of snake (Walterinnesia aegyptia) venom-loaded silica nanoparticles in the treatment of breast cancer- and prostate cancer-bearing experimental mouse models. Free Radic Biol Med. 65:175–189. 2013. View Article : Google Scholar : PubMed/NCBI

249 

An S, Tiruthani K, Wang Y, Xu L, Hu M, Li J, Song W, Jiang H, Sun J, Liu R and Huang L: Locally trapping the C-C chemokine receptor type 7 by gene delivery nanoparticle inhibits lymphatic metastasis prior to tumor resection. Small. 15:e18051822019. View Article : Google Scholar : PubMed/NCBI

250 

Liu JQ, Zhang C, Zhang X, Yan J, Zeng C, Talebian F, Lynch K, Zhao W, Hou X, Du S, et al: Intratumoral delivery of IL-12 and IL-27 mRNA using lipid nanoparticles for cancer immunotherapy. J Control Release. 345:306–313. 2022. View Article : Google Scholar : PubMed/NCBI

251 

Li Y, Su Z, Zhao W, Zhang X, Momin N, Zhang C, Wittrup KD, Dong Y, Irvine DJ and Weiss R: Multifunctional oncolytic nanoparticles deliver self-replicating IL-12 RNA to eliminate established tumors and prime systemic immunity. Nat Cancer. 1:882–893. 2020. View Article : Google Scholar

252 

Nguyen KG, Vrabel MR, Mantooth SM, Hopkins JJ, Wagner ES, Gabaldon TA and Zaharoff DA: Localized interleukin-12 for cancer immunotherapy. Front Immunol. 11:5755972020. View Article : Google Scholar : PubMed/NCBI

253 

Xu S, Xu Y, Solek NC, Chen J, Gong F, Varley AJ, Golubovic A, Pan A, Dong S, Zheng G and Li B: Tumor-tailored ionizable lipid nanoparticles facilitate IL-12 circular RNA delivery for enhanced lung cancer immunotherapy. Adv Mater. 36:e24003072024. View Article : Google Scholar : PubMed/NCBI

254 

Fu S, Li G, Zang W, Zhou X, Shi K and Zhai Y: Pure drug nano-assemblies: A facile carrier-free nanoplatform for efficient cancer therapy. Acta Pharm Sin B. 12:92–106. 2022. View Article : Google Scholar : PubMed/NCBI

255 

Reda M, Ngamcherdtrakul W, Nelson MA, Siriwon N, Wang R, Zaidan HY, Bejan DS, Reda S, Hoang NH, Crumrine NA, et al: Development of a nanoparticle-based immunotherapy targeting PD-L1 and PLK1 for lung cancer treatment. Nat Commun. 13:42612022. View Article : Google Scholar : PubMed/NCBI

256 

Han X, Wei Q, Lv Y, Weng L, Huang H, Wei Q, Li M, Mao Y, Hua D, Cai X, et al: Ginseng-derived nanoparticles potentiate immune checkpoint antibody efficacy by reprogramming the cold tumor microenvironment. Mol Ther. 30:327–340. 2022. View Article : Google Scholar :

257 

Liu L, Wang Y, Miao L, Liu Q, Musetti S, Li J and Huang L: Combination immunotherapy of MUC1 mRNA nano-vaccine and CTLA-4 blockade effectively inhibits growth of triple negative breast cancer. Mol Ther. 26:45–55. 2018. View Article : Google Scholar :

258 

Ma R, Li Z, Chiocca EA, Caligiuri MA and Yu J: The emerging field of oncolytic virus-based cancer immunotherapy. Trends Cancer. 9:122–139. 2023. View Article : Google Scholar :

259 

Hu H, Zhang S, Cai L, Duan H, Li Y, Yang J, Wang Y and Liu B, Dong S, Fang Z and Liu B: A novel cocktail therapy based on quintuplet combination of oncolytic herpes simplex virus-2 vectors armed with interleukin-12, interleukin-15, GM-CSF, PD1v, and IL-7 × CCL19 results in enhanced antitumor efficacy. Virol J. 19:742022. View Article : Google Scholar

260 

Kim KJ, Moon D, Kong SJ, Lee YS, Yoo Y, Kim S, Kim C, Chon HJ, Kim JH and Choi KJ: Antitumor effects of IL-12 and GM-CSF co-expressed in an engineered oncolytic HSV-1. Gene Ther. 28:186–198. 2021. View Article : Google Scholar

261 

Oh E, Oh JE, Hong J, Chung Y, Lee Y, Park KD, Kim S and Yun CO: Optimized biodegradable polymeric reservoir-mediated local and sustained co-delivery of dendritic cells and oncolytic adenovirus co-expressing IL-12 and GM-CSF for cancer immunotherapy. J Control Release. 259:115–127. 2017. View Article : Google Scholar : PubMed/NCBI

262 

Zhang SN, Choi IK, Huang JH, Yoo JY, Choi KJ and Yun CO: Optimizing DC vaccination by combination with oncolytic adenovirus coexpressing IL-12 and GM-CSF. Mol Ther. 19:1558–1568. 2011. View Article : Google Scholar : PubMed/NCBI

263 

Malhotra J and Kim ES: Oncolytic viruses and cancer immunotherapy. Curr Oncol Rep. 25:19–28. 2023. View Article : Google Scholar

Related Articles

Journal Cover

July-2025
Volume 67 Issue 1

Print ISSN: 1019-6439
Online ISSN:1791-2423

Sign up for eToc alerts

Recommend to Library

Copy and paste a formatted citation
x
Spandidos Publications style
Lyu X, Han J, Lin C, Zhou Y and Wang W: Beyond the tumor microenvironment: Orchestrating systemic T‑cell response for next‑generation cancer immunotherapy (Review). Int J Oncol 67: 56, 2025.
APA
Lyu, X., Han, J., Lin, C., Zhou, Y., & Wang, W. (2025). Beyond the tumor microenvironment: Orchestrating systemic T‑cell response for next‑generation cancer immunotherapy (Review). International Journal of Oncology, 67, 56. https://doi.org/10.3892/ijo.2025.5762
MLA
Lyu, X., Han, J., Lin, C., Zhou, Y., Wang, W."Beyond the tumor microenvironment: Orchestrating systemic T‑cell response for next‑generation cancer immunotherapy (Review)". International Journal of Oncology 67.1 (2025): 56.
Chicago
Lyu, X., Han, J., Lin, C., Zhou, Y., Wang, W."Beyond the tumor microenvironment: Orchestrating systemic T‑cell response for next‑generation cancer immunotherapy (Review)". International Journal of Oncology 67, no. 1 (2025): 56. https://doi.org/10.3892/ijo.2025.5762