Skip to main content

Main menu

  • Home
  • About
    • About CBM
    • Editorial Board
    • Announcement
  • Articles
    • Ahead of print
    • Current Issue
    • Archive
    • Collections
    • Cover Story
  • For Authors
    • Instructions for Authors
    • Resources
    • Submit a Manuscript
  • For Reviewers
    • Become a Reviewer
    • Instructions for Reviewers
    • Resources
    • Outstanding Reviewer
  • Subscription
  • Alerts
    • Email Alerts
    • RSS Feeds
    • Table of Contents
  • Contact us
  • Other Publications
    • cbm

User menu

  • My alerts

Search

  • Advanced search
Cancer Biology & Medicine
  • Other Publications
    • cbm
  • My alerts
Cancer Biology & Medicine

Advanced Search

 

  • Home
  • About
    • About CBM
    • Editorial Board
    • Announcement
  • Articles
    • Ahead of print
    • Current Issue
    • Archive
    • Collections
    • Cover Story
  • For Authors
    • Instructions for Authors
    • Resources
    • Submit a Manuscript
  • For Reviewers
    • Become a Reviewer
    • Instructions for Reviewers
    • Resources
    • Outstanding Reviewer
  • Subscription
  • Alerts
    • Email Alerts
    • RSS Feeds
    • Table of Contents
  • Contact us
  • Follow cbm on Twitter
  • Visit cbm on Facebook
EditorialEditorial
Open Access

PD-1 and LAG-3 dual blockade: emerging mechanisms and potential therapeutic prospects in cancer

Xiangyu Qiu, Zhaoan Yu, Xiaoqing Lu, Xin Jin, Jinrong Zhu and Rongxin Zhang
Cancer Biology & Medicine December 2024, 20240436; DOI: https://doi.org/10.20892/j.issn.2095-3941.2024.0436
Xiangyu Qiu
Guangdong Provincial Key Laboratory of Advanced Drug Delivery, Guangdong Provincial Engineering Center of Topical Precise Drug Delivery System, School of Life Sciences and Biopharmaceutics, Guangdong Pharmaceutical University, Guangzhou 510006, China
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
Zhaoan Yu
Guangdong Provincial Key Laboratory of Advanced Drug Delivery, Guangdong Provincial Engineering Center of Topical Precise Drug Delivery System, School of Life Sciences and Biopharmaceutics, Guangdong Pharmaceutical University, Guangzhou 510006, China
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
Xiaoqing Lu
Guangdong Provincial Key Laboratory of Advanced Drug Delivery, Guangdong Provincial Engineering Center of Topical Precise Drug Delivery System, School of Life Sciences and Biopharmaceutics, Guangdong Pharmaceutical University, Guangzhou 510006, China
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
Xin Jin
Guangdong Provincial Key Laboratory of Advanced Drug Delivery, Guangdong Provincial Engineering Center of Topical Precise Drug Delivery System, School of Life Sciences and Biopharmaceutics, Guangdong Pharmaceutical University, Guangzhou 510006, China
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
Jinrong Zhu
Guangdong Provincial Key Laboratory of Advanced Drug Delivery, Guangdong Provincial Engineering Center of Topical Precise Drug Delivery System, School of Life Sciences and Biopharmaceutics, Guangdong Pharmaceutical University, Guangzhou 510006, China
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
Rongxin Zhang
Guangdong Provincial Key Laboratory of Advanced Drug Delivery, Guangdong Provincial Engineering Center of Topical Precise Drug Delivery System, School of Life Sciences and Biopharmaceutics, Guangdong Pharmaceutical University, Guangzhou 510006, China
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
  • ORCID record for Rongxin Zhang
  • For correspondence: zhujinrong{at}gdpu.edu.cn rxzhang{at}gdpu.edu.cn
  • Article
  • Figures & Data
  • Info & Metrics
  • References
  • PDF
Loading

The emergence of tumor immunotherapy represents a pivotal milestone in cancer treatment, heralding new hope for cancer patients globally. Immune checkpoint inhibitors have been instrumental in this advance, playing a crucial role in modulating the immune system response to neoplasms. Specifically, the therapies targeting the programmed death 1/programmed cell death ligand 1 (PD-1/PD-L1) immune checkpoint pathways have shown significant efficacy in treating various cancers1. Although PD-1/PD-L1 inhibitors have achieved considerable success in the treatment of cancer, the effectiveness of PD-1/PD-L1 inhibitors as single-agent therapy is currently constrained. In recent years strategies to block multiple immune checkpoints simultaneously have received a lot of attention2. Currently, dual blockade strategies targeting PD-1 and lymphocyte activation gene 3 (LAG-3) are gaining increased attention due to the significant potential to enhance anti-tumor immune responses and becoming a focal point in cancer research. Emerging mechanisms in this domain and insights into potential therapeutic prospects are discussed in this editorial. Additionally, recent progress in the challenges and clinical application of PD-1 and LAG-3 dual blockade is discussed.

Synergistic anti-tumor mechanism underlying anti-LAG-3 and anti-PD-1

PD-1 has been a key immune checkpoint in cancer immunotherapy. PD-1 has an important role in regulating the immune response and preventing an overactive immune system by attenuating T-cell activity through interaction with the ligands, PD-L1 or PD-L23. The underlying mechanism is integral to the maintenance of immune homeostasis. Nevertheless, malignant cells frequently utilize the PD-1 pathway to evade immune surveillance, thereby avoiding detection and elimination by the host immune system. Therefore, PD-1 inhibitors, such as nivolumab and pembrolizumab, were developed to block the related signaling pathway by tumor cells and restore the capacity of the immune system to recognize and attack tumor cells. Clinical evidence has confirmed the effectiveness of PD-1 inhibitors4. PD-1 checkpoint therapy has progressed significantly but still faces notable limitations. PD-1 inhibitors are not universally effective for all patients. Additionally, the occurrence of immune-related adverse events, such as rashes and endocrine disorders, remains a significant issue requiring attention and resolution.

Considering the limitations and side effects of PD-1 blockade therapy, researchers are exploring other immune checkpoints to improve therapeutic effects. LAG-3 has received considerable interest as a promising new target. LAG-3 is an inhibitory receptor primarily expressed on exhausted T cells. LAG-3 inhibits T cell activation and function by interacting with MHC class II molecules and other ligands, including galectin-3 (Gal-3), fibrinogen-like protein 1 (FGL1), and liver and lymph node sinusoidal endothelial cell C-type lectin (LSECtin)5. Upregulation of LAG-3 leads to T-cell exhaustion, thereby promoting tumor immune escape. LAG-3 has a critical role in T cells and is also expressed in other immune cell types, including regulatory T and natural killer cells6. Clinical trials have demonstrated the significant efficacy of LAG-3 blockers in isolation or in conjunction with other immune checkpoint inhibitors (ICIs), indicating the potential to improve patient prognosis and enhance the immune system response7.

Studies have shown that PD-1 and LAG-3 are co-expressed in the tumor immune microenvironment, jointly mediating the immune escape of cancer cells8. When expressed simultaneously, PD-1 and LAG-3 synergistically inhibit T cell activation signaling through distinct but complementary pathways and activate T cell exhaustion signaling pathways. T-cell exhaustion refers to the gradual decline in T cell function within the immune system, a phenomenon influenced by various factors, including persistent antigen exposure, the impact of immunosuppressive signals, the role of soluble mediators, and the expression levels of multiple transcription factors associated with T-cell exhaustion (Figure 1). Vignali and colleagues discovered that CD8+ T cells with dual PD-1 and LAG-3 blockade exhibited stronger tumor clearance ability and long-term survival in a melanoma mouse model compared to CD8+ T cell blockade with either receptor alone. These dual-blockade CD8+ T cells also displayed extensive T cell receptor (TCR) clonality and enrichment of effector-like and interferon-responsive genes, resulting in increased release of IFN-γ9. Another clinical trial (NCT03743766) revealed the complex molecular mechanisms underlying combined LAG-3 and PD-1 blockade. The study showed that combination therapy (relatlimab and nivolumab) was safe and effective, enhancing the CD8+ T cell receptor signaling capacity, lowering the threshold for TCR activation, and altering CD8+ T cell differentiation to increase cytotoxicity while retaining exhaustion characteristics10. This research demonstrated that cytotoxicity can be enhanced while preserving exhaustion features, guiding future immunotherapy strategies. Concurrently, Wherry et al. reported that during chronic viral infections and cancer, exhausted CD8+ T cells persistently co-express inhibitory receptors with PD-1 and LAG-3 having distinct and complementary roles in CD8+ T cell exhaustion. PD-1 primarily inhibits T cell proliferation, while LAG-3 regulates T cell cytotoxicity and cytokine production. Blocking PD-1 and LAG-3 reactivates T exhaustion (Tex) cells. This finding provided new insights into how LAG-3 and PD-1 promote T-cell exhaustion differently11.

Figure 1
  • Download figure
  • Open in new tab
  • Download powerpoint
Figure 1

The causes leading to T-cell exhaustion. Ⅰ. Persistent antigen exposure: persistent antigen exposure due to chronic infection or cancer. The production of type I interferon (IFN α/β) continues to increase and the high level of IFN α/β signaling affects T-cell function, promoting T-cell exhaustion. Ⅱ. Inhibitory receptors: inhibitory receptors on the surface of T cells, such as PD-1, LAG-3, and CTLA-4, bind to the corresponding ligands on the surface of the tumor, resulting in inhibition of T-cell activation, reduction of cytokine production, and weakening of T-cell function, which leads to T-cell exhaustion. Ⅲ. Soluble mediators: soluble molecules are a second class of signals that regulate T-cell exhaustion, including immunosuppressive cytokines, such as IL-10 and transforming growth factor-beta (TGF-β), and inflammatory cytokines, such as IFN-α and IL-6. Ⅳ. Transcription factors associated with T-cell exhaustion: PR domain containing 1 (PRMD1), basic leucine zipper ATF-like transcription factor (BATF), ETS variant transcription factor 7 (ETV7), and thymocyte selection-associated high mobility group box (TOX) are transcription factors, closely associated with T-cell exhaustion. Transcription factors associated with T-cell exhaustion have an important role in regulating the CD8+ T-cell function and exhaustion state. The expression of transcription factors is elevated in exhausted T cells.

In summary, LAG-3 and PD-1 synergistically regulate T cell exhaustion through non-redundant mechanisms, offering new strategies for clinical anti-tumor guidance (Figure 2).

Figure 2
  • Download figure
  • Open in new tab
  • Download powerpoint
Figure 2

The impact of the PD-1 and LAG-3 pathways on a tumor cell in blocked and unblocked states. Major histocompatibility complex (MHC) molecules and PD-L1 on the surface of tumor cells and antigen-presenting cells (APCs) interact with the inhibitory receptors LAG-3 and PD-1 on T cells, suppressing T-cell activation. By activating the PD-1 and LAG-3 immunosuppressive pathways, these immune checkpoints promote the development of exhausted T cells by inhibiting proliferation, cytokine production (IFN-γ), and cytotoxicity functions. Additionally, enhancing the expression of T-cell exhaustion-associated transcription factors, such as TOX, leads to tumor immune escape. After treatment with relatlimab and nivolumab, which effectively block the activation of the PD-1 and LAG-3 signaling pathways, the combination of immunosuppressants enhances T-cell receptor (TCR) signaling in CD8+ T cells and alters the differentiation of CD8+ T cells, restoring the proliferative capacity of T cells. Inhibition of the expression of T-cell exhaustion-related transcription factors such as TOX, simultaneously increased the release of IFN-γ and enhanced anti-tumor immunity, resulting in the effective elimination of tumor cells.

Clinical advances in combination therapy for cancer

The combination of PD-1 and LAG-3 have shown promising prospects in clinical trials. Notably, Bristol Myers Squibb developed a fixed-dose combination immunotherapy with relatlimab (anti-LAG-3 antibody) and nivolumab (anti-PD-1 antibody)12. In a phase III clinical trial (RELATIVITY-047) involving patients with advanced melanoma, this combination significantly extended progression-free survival (PFS) from a median of 4.6 months to 10.1 months compared to nivolumab alone13. Based on these results, the U.S. FDA approved this combination for treating advanced melanoma in March 2022, becoming the first approved LAG-3-targeted therapy.

To validate the efficacy and breadth of this dual blockade immunotherapy, the dual blockade strategy of PD-1 and LAG-3 is gradually being tested in clinical trials for other solid tumors. In a study involving 60 patients with resectable non-small cell lung cancer, 95% achieved complete surgical resection and 12-month disease-free and overall survival (OS) rates reached 93% and 100%, respectively, following combination treatment with nivolumab and relatlimab14. In patients with metastatic head and neck squamous cell carcinoma, combination treatment with eftilagimod alpha (soluble LAG-3 protein) and pembrolizumab (PD-1 antagonist) achieved an objective response rate of 29.7% with 13.5% of patients achieving complete responses and 80% of confirmed responders maintaining response after 12 months. This result was a significant improvement compared to ICIs alone15. The combination of favezelimab (LAG-3 monoclonal antibody) and pembrolizumab (PD-1 monoclonal antibody) resulted in higher response rates and prolonged survival in patients with microsatellite-stable advanced colorectal cancer with PD-L1 CPS ≥ 116. The combination of nivolumab and relatlimab achieved an objective response rate of 50% and PFS of 27.5% in patients with microsatellite instability-high/mismatch repair deficient (MSI-H/dMMR) metastatic colorectal cancer, providing durable clinical benefits and manageable safety for colorectal cancer patients17.

Multifaceted clinical trials indicate that combining anti-PD-1 and anti-LAG-3 therapies benefit patients who do not respond to traditional immunotherapy. Scientists have broadened the scope of immunotherapy while advancing personalized and precision treatments using this approach across various cancer types (Table 1).

View this table:
  • View inline
  • View popup
Table 1

Summary of clinical trials involving PD-1 and LAG-3 single or combination immune checkpoint inhibitor treatment

Challenges of combination therapy

Combining PD-1 and LAG-3 inhibitors can improve cancer patient outcomes, advance immunotherapy, and bring new hope for survival. However, this combination therapy also faces some challenges.

Mechanism to be elucidated

PD-1 and LAG-3 are critical immune checkpoint molecules that regulate the activity of the immune system through different mechanisms but the signaling pathways may cross or interact within some cells. To gain insight into combination inhibitory therapies with PD-1 and LAG-3 and to improve the efficacy of tumor immunotherapy, an in-depth study of the mechanisms underlying the interactions is essential. Currently, the understanding of the specific mechanisms of action of PD-1 and LAG-3 and the interactions in multiple immune cells is incomplete. Therefore, research must be intensive to elucidate the complex biological functions in different immune cell populations.

Increased immune adverse reactions

Dual inhibition of immune checkpoints may lead to stronger immune system activation and cause higher immune-related adverse events (irAEs). The combination of relatlimab and nivolumab resulted in a higher proportion of grade 3 or 4 treatment-related adverse events (18.9%) in the RELATIVITY-047 study compared to 9.7% of patients treated with nivolumab alone. Three deaths in the combination treatment group were treatment-related compared to two deaths in the monotherapy group13. Another study in patients with advanced gastric and gastroesophageal junction cancers showed that the rate of grade 3 or 4 treatment-related adverse events was slightly higher in the nivolumab + relatlimab + chemotherapy group (69%) than the nivolumab + chemotherapy group (61%) and more patients discontinued the medication due to adverse events18. This regimen requires careful patient monitoring and dose optimization to balance effectiveness and safety.

Scope of tumor application to be expanded

Different tumor types may respond differently to this combination therapy. Some tumors may be more dependent on a particular immunosuppressive pathway. Although dual blockade combination therapy with PD-1 and LAG-3 has shown better efficacy in a variety of solid tumors, dual blockade combination therapy with PD-1 and LAG-3 is slightly less effective than monotherapy in advanced gastric cancer. The addition of relatlimab did not significantly improve the objective remission rate (ORR) in patients with LAG-3 expression in ≥ 1% of tumors in the RELATIVITY-060 phase II study and PFS and OS were not significantly improved compared to nivolumab and chemotherapy alone18. Therefore, the scope of this strategy needs to be clarified in clinical trials.

Exploring future research directions in combination therapy

To enhance the effectiveness of LAG-3 and PD-1 inhibition in cancer treatment, further investigation is required to elucidate the following aspects.

More specific mechanism research

To maximize the utilization of LAG-3 and PD-1 inhibition in cancer treatment, it is essential to analyze the specific mechanisms underlying immune regulation. Specifically, revealing the LAG-3 and PD-1 expression patterns and functions in specific types of immune cells (T, natural killer, and dendritic cells) may discover how LAG-3 and PD-1 synergistically or separately suppress immune responses. Studying the variations in these signals within different tumor microenvironments, such as oxygen-rich and oxygen-poor conditions, can also provide important insights for developing new strategies and precisely targeted interventions to improve the specificity and efficacy of therapies.

Developing more reliable biomarkers to predict the response to combination therapy

Identifying and validating biomarkers that can predict patient response to LAG-3 and PD-1 combination therapy in cancer immunotherapy is crucial for achieving personalized treatment strategies. Ideal biomarkers should evaluate efficacy and potential adverse reactions early in treatment, enabling targeted adjustments to the treatment plan.

Exploration of more treatment combinations

In addition to LAG-3, T cell immunoglobulin and mucin domain-containing protein 3 (TIM-3 T) and T cell immunoglobulin and ITIM domains (TIGIT) are emerging as promising targets, all of which have demonstrated potential in treating tumors. Investigation multiple immune checkpoint therapy combination, as well as the effects resulting from the combination of ICIs with small-molecule drugs, chemotherapy, or radiotherapy, may offer opportunities for enhancing efficacy and overcoming the limitations associated with single-modality treatments. These multifaceted therapeutic strategies are poised to make significant advances in tumor treatment.

Research on resistance mechanisms

Tumors can develop resistance through gene mutations, signaling pathway reprogramming, immune evasion, and alterations in the tumor microenvironment. Additionally, changes in inhibitory cytokines and metabolic environments within the tumor microenvironment can weaken the activity of immunotherapy. Even though current immune checkpoint inhibition therapies show significant efficacy in some patients, tumors frequently develop resistance through multiple mechanisms. This represents a significant challenge because the emergence of resistance has the effect of reducing the effectiveness of treatment and limiting the survival time of patients. Therefore, studying resistance pathways and mechanisms and identifying causes of treatment failure is key to improving therapy durability and patient quality of life.

Exploring the potential of combination therapy in other tumor treatments to expand the range of indications

Although current research mainly focuses on certain types of cancer, exploring the application potential in different tissue types and tumor markers may expand the range of applicable tumors. By conducting cross-tumor studies and clinical trials, the potential advantages of combination therapy in various cancers may be demonstrated.

Discussion

The combined inhibition strategy of PD-1 and LAG-3 has shown significant potential in current cancer immunotherapy research, especially in enhancing anti-tumor immune responses and improving patient prognosis. Clinical trials have demonstrated that this combination therapy has achieved positive results in some cancer types. For example, the fixed-dose combination of relatlimab (anti-LAG-3 antibody) and nivolumab (anti-PD-1 antibody) developed by Bristol Myers Squibb significantly extended PFS from a median of 4.6 months to 10.1 months in patients with advanced melanoma. Additionally, studies for other cancer types, such as non-small cell lung cancer and metastatic head and neck squamous cell carcinoma, have also shown promising prospects. Although clinical trials have demonstrated the positive effects of this combination therapy in certain cancer types, further investigation into the underlying mechanisms and the range of indications in other tumor types is necessary. The roles of PD-1 and LAG-3 in regulating T cell exhaustion provide new therapeutic insights, but the specific signaling pathway interactions remain to be fully elucidated. Hence, forthcoming research should focus on revealing the expression patterns and functions of these two immune checkpoints in various immune cells, especially the alterations in different tumor microenvironments. Moreover, developing reliable biomarkers to predict patient response to combination therapy will facilitate personalized treatment strategies. By identifying biomarkers to evaluate efficacy and potential adverse reactions at the onset of treatment, physicians can more effectively tailor treatment plans to improve patient outcomes and quality of life. By conducting cross-tumor studies and clinical trials we can identify the potential advantages of combination therapy in different cancers, ultimately benefiting more patients. In conclusion, despite the challenges faced by combination therapy, the prospects remain promising and deserve in-depth study.

Conflict of interest statement

No potential conflicts of interest are disclosed.

Author contributions

Conceived and designed the analysis: Jinrong Zhu, Rongxin Zhang.

Wrote the paper: Xiangyu Qiu.

Collected the data: Zhaoan Yu, Xiaoqing Lu, Xin Jin.

  • Received October 5, 2024.
  • Accepted November 6, 2024.
  • Copyright: © 2024, The Authors

This work is licensed under the Creative Commons Attribution-NonCommercial 4.0 International License.

References

  1. 1.↵
    1. Lin X,
    2. Kang K,
    3. Chen P,
    4. Zeng Z,
    5. Li G,
    6. Xiong W, et al.
    Regulatory mechanisms of PD-1/PD-L1 in cancers. Mol Cancer. 2024; 23: 108.
    OpenUrlPubMed
  2. 2.↵
    1. Patel SA,
    2. Minn AJ.
    Combination cancer therapy with immune checkpoint blockade: mechanisms and strategies. Immunity. 2018; 48: 417–33.
    OpenUrlPubMed
  3. 3.↵
    1. Kong X,
    2. Zhang J,
    3. Chen S,
    4. Wang X,
    5. Xi Q,
    6. Shen H, et al.
    Immune checkpoint inhibitors: breakthroughs in cancer treatment. Cancer Biol Med. 2024; 21: 451–72.
    OpenUrlAbstract/FREE Full Text
  4. 4.↵
    1. Jia L,
    2. Zhang Q,
    3. Zhang R.
    PD-1/PD-L1 pathway blockade works as an effective and practical therapy for cancer immunotherapy. Cancer Biol Med. 2018; 15: 116–23.
    OpenUrlAbstract/FREE Full Text
  5. 5.↵
    1. Aggarwal V,
    2. Workman CJ,
    3. Vignali DAA.
    LAG-3 as the third checkpoint inhibitor. Nat Immunol. 2023; 24: 1415–22.
    OpenUrlPubMed
  6. 6.↵
    1. Huuhtanen J,
    2. Kasanen H,
    3. Peltola K,
    4. Lonnberg T,
    5. Glumoff V,
    6. Bruck O, et al.
    Single-cell characterization of anti-LAG-3 and anti-PD-1 combination treatment in patients with melanoma. J Clin Invest. 2023; 133: e164809.
  7. 7.↵
    1. Schoffski P,
    2. Tan DSW,
    3. Martin M,
    4. Ochoa-de-Olza M,
    5. Sarantopoulos J,
    6. Carvajal RD, et al.
    Phase I/II study of the LAG-3 inhibitor ieramilimab (LAG525) +/− anti-PD-1 spartalizumab (PDR001) in patients with advanced malignancies. J Immunother Cancer. 2022; 10: e003776.
  8. 8.↵
    1. Woo SR,
    2. Turnis ME,
    3. Goldberg MV,
    4. Bankoti J,
    5. Selby M,
    6. Nirschl CJ, et al.
    Immune inhibitory molecules LAG-3 and PD-1 synergistically regulate T-cell function to promote tumoral immune escape. Cancer Res. 2012; 72: 917–27.
    OpenUrlAbstract/FREE Full Text
  9. 9.↵
    1. Andrews LP,
    2. Butler SC,
    3. Cui J,
    4. Cillo AR,
    5. Cardello C,
    6. Liu C, et al.
    LAG-3 and PD-1 synergize on CD8(+) T cells to drive T cell exhaustion and hinder autocrine IFN-gamma-dependent anti-tumor immunity. Cell. 2024; 187: 4355–72.e22.
    OpenUrlPubMed
  10. 10.↵
    1. Cillo AR,
    2. Cardello C,
    3. Shan F,
    4. Karapetyan L,
    5. Kunning S,
    6. Sander C, et al.
    Blockade of LAG-3 and PD-1 leads to co-expression of cytotoxic and exhaustion gene modules in CD8(+) T cells to promote antitumor immunity. Cell. 2024; 187: 4373–88.e15.
    OpenUrlPubMed
  11. 11.↵
    1. Ngiow SF,
    2. Manne S,
    3. Huang YJ,
    4. Azar T,
    5. Chen Z,
    6. Mathew D, et al.
    LAG-3 sustains TOX expression and regulates the CD94/NKG2-Qa-1b axis to govern exhausted CD8 T cell NK receptor expression and cytotoxicity. Cell. 2024; 187: P4336–54.E19.
    OpenUrl
  12. 12.↵
    1. Paik J.
    Nivolumab plus relatlimab: first approval. Drugs. 2022; 82: 925–31.
    OpenUrlPubMed
  13. 13.↵
    1. Tawbi HA,
    2. Schadendorf D,
    3. Lipson EJ,
    4. Ascierto PA,
    5. Matamala L,
    6. Castillo Gutierrez E, et al.
    Relatlimab and nivolumab versus nivolumab in untreated advanced melanoma. N Engl J Med. 2022; 386: 24–34.
    OpenUrlCrossRefPubMed
  14. 14.↵
    1. Schuler M,
    2. Cuppens K,
    3. Plones T,
    4. Wiesweg M,
    5. Du Pont B,
    6. Hegedus B, et al.
    Neoadjuvant nivolumab with or without relatlimab in resectable non-small-cell lung cancer: a randomized phase 2 trial. Nat Med. 2024; 30: 1602–11.
    OpenUrlPubMed
  15. 15.↵
    1. Forster M,
    2. Brana I,
    3. Pousa AL,
    4. Doger B,
    5. Roxburgh P,
    6. Bajaj P, et al.
    Eftilagimod alpha (soluble LAG3 protein) combined with pembrolizumab as second-line therapy for patients with metastatic head and neck squamous cell carcinoma. Clin Cancer Res. 2024; 30: 3726–34.
    OpenUrlPubMed
  16. 16.↵
    1. Garralda E,
    2. Sukari A,
    3. Lakhani NJ,
    4. Patnaik A,
    5. Lou Y,
    6. Im SA, et al.
    A first-in-human study of the anti-LAG-3 antibody favezelimab plus pembrolizumab in previously treated, advanced microsatellite stable colorectal cancer. ESMO Open. 2022; 7: 100639.
  17. 17.↵
    1. Overman MJ,
    2. Gelsomino F,
    3. Aglietta M,
    4. Wong M,
    5. Limon Miron ML,
    6. Leonard G, et al.
    Nivolumab plus relatlimab in patients with previously treated microsatellite instability-high/mismatch repair-deficient metastatic colorectal cancer: the phase II CheckMate 142 study. J Immunother Cancer. 2024; 12: e008689.
  18. 18.↵
    1. Hegewisch-Becker S,
    2. Mendez G,
    3. Chao J,
    4. Nemecek R,
    5. Feeney K,
    6. Van Cutsem E, et al.
    First-line nivolumab and relatlimab plus chemotherapy for gastric or gastroesophageal junction adenocarcinoma: the phase II RELATIVITY-060 study. J Clin Oncol. 2024; 42: 2080–93.
    OpenUrlPubMed
Next
Back to top

In this issue

Cancer Biology & Medicine: 22 (10)
Cancer Biology & Medicine
Vol. 22, Issue 10
15 Oct 2025
  • Table of Contents
  • Index by author
Print
Download PDF
Email Article

Thank you for your interest in spreading the word on Cancer Biology & Medicine.

NOTE: We only request your email address so that the person you are recommending the page to knows that you wanted them to see it, and that it is not junk mail. We do not capture any email address.

Enter multiple addresses on separate lines or separate them with commas.
PD-1 and LAG-3 dual blockade: emerging mechanisms and potential therapeutic prospects in cancer
(Your Name) has sent you a message from Cancer Biology & Medicine
(Your Name) thought you would like to see the Cancer Biology & Medicine web site.
Citation Tools
PD-1 and LAG-3 dual blockade: emerging mechanisms and potential therapeutic prospects in cancer
Xiangyu Qiu, Zhaoan Yu, Xiaoqing Lu, Xin Jin, Jinrong Zhu, Rongxin Zhang
Cancer Biology & Medicine Dec 2024, 20240436; DOI: 10.20892/j.issn.2095-3941.2024.0436

Citation Manager Formats

  • BibTeX
  • Bookends
  • EasyBib
  • EndNote (tagged)
  • EndNote 8 (xml)
  • Medlars
  • Mendeley
  • Papers
  • RefWorks Tagged
  • Ref Manager
  • RIS
  • Zotero
Share
PD-1 and LAG-3 dual blockade: emerging mechanisms and potential therapeutic prospects in cancer
Xiangyu Qiu, Zhaoan Yu, Xiaoqing Lu, Xin Jin, Jinrong Zhu, Rongxin Zhang
Cancer Biology & Medicine Dec 2024, 20240436; DOI: 10.20892/j.issn.2095-3941.2024.0436
Twitter logo Facebook logo Mendeley logo
  • Tweet Widget
  • Facebook Like
  • Google Plus One

Jump to section

  • Article
    • Synergistic anti-tumor mechanism underlying anti-LAG-3 and anti-PD-1
    • Clinical advances in combination therapy for cancer
    • Challenges of combination therapy
    • Exploring future research directions in combination therapy
    • Discussion
    • Conflict of interest statement
    • Author contributions
    • References
  • Figures & Data
  • Info & Metrics
  • References
  • PDF

Related Articles

  • No related articles found.
  • Google Scholar

Cited By...

  • No citing articles found.
  • Google Scholar

More in this TOC Section

  • Chimeric antigen receptor macrophages: a new frontier in hepatocellular carcinoma treatment
  • Advances in antibody-drug conjugates for urothelial carcinoma treatment
  • Progress toward cervical cancer elimination: global disparities and China’s contributions
Show more Editorial

Similar Articles

Navigate

  • Home
  • Current Issue

More Information

  • About CBM
  • About CACA
  • About TMUCIH
  • Editorial Board
  • Subscription

For Authors

  • Instructions for authors
  • Journal Policies
  • Submit a Manuscript

Journal Services

  • Email Alerts
  • Facebook
  • RSS Feeds
  • Twitter

 

© 2025 Cancer Biology & Medicine

Powered by HighWire