Next Article in Journal
Determination of the Potential Tumor-Suppressive Effects of Gsdme in a Chemically Induced and in a Genetically Modified Intestinal Cancer Mouse Model
Previous Article in Journal
Circulating Tumor Cell Enumeration and Characterization in Metastatic Castration-Resistant Prostate Cancer Patients Treated with Cabazitaxel
Previous Article in Special Issue
Immunotherapy in Hodgkin Lymphoma: Present Status and Future Strategies
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

LAG3: The Biological Processes That Motivate Targeting This Immune Checkpoint Molecule in Human Cancer

by
Cinzia Solinas
1,2,
Edoardo Migliori
1,3,
Pushpamali De Silva
1 and
Karen Willard-Gallo
1,*
1
Molecular Immunology Unit, Institut Jules Bordet, Université Libre de Bruxelles, 1000 Brussels, Belgium
2
Azienda Unità Sanitaria Locale Valle d’Aosta, Regional Hospital of Aosta, 11100 Aosta, Italy
3
Columbia University Medical Center, Columbia Center for Translational Immunology, NY 10032, USA
*
Author to whom correspondence should be addressed.
Submission received: 22 July 2019 / Revised: 12 August 2019 / Accepted: 14 August 2019 / Published: 20 August 2019
(This article belongs to the Special Issue Signaling Pathways and Immune Checkpoint Regulation in Cancer)

Abstract

:
The programmed cell death 1 (PD-1) pathway is an important regulator of immune responses in peripheral tissues, including abnormal situations such as the tumor microenvironment. This pathway is currently the principal target for immunotherapeutic compounds designed to block immune checkpoint pathways, with these drugs improving clinical outcomes in a number of solid and hematological tumors. Medical oncology is experiencing an immune revolution that has scientists and clinicians looking at alternative, non-redundant inhibitory pathways also involved in regulating immune responses in cancer. A variety of targets have emerged for combinatorial approaches in immune checkpoint blockade. The main purpose of this narrative review is to summarize the biological role of lymphocyte activation gene 3 (LAG3), an emerging targetable inhibitory immune checkpoint molecule. We briefly discuss its role in infection, autoimmune disease and cancer, with a more detailed analysis of current data on LAG3 expression in breast cancer. Current clinical trials testing soluble LAG3 immunoglobulin and LAG3 antagonists are also presented in this work.

1. Introduction

Improved clinical outcomes have been achieved for a number of solid and hematological diseases treated with immune checkpoint blockade (ICB) targeting cytotoxic T lymphocyte associated protein (CTLA)-4 and programmed cell death 1 (PD-1) or its ligand PD-L1 [1,2,3,4,5,6,7,8,9,10,11]. Nevertheless, a large proportion of ICB-treated cancer patients still do not benefit from these drugs. Thus, while initial ICB targets have led to an immunological resurgence in oncology, this lack of widespread clinical benefit together with the occurrence of immune related adverse events (irAEs), principally due to the onset of autoimmune reactions [12,13,14,15,16,17], have focused attention on alternative inhibitory immune checkpoint molecules, including lymphocyte activation gene 3 (LAG3, CD223), T cell immunoglobulin mucin 3 (TIM3) [18], adenosine A2A receptor (A2AR) [19], indoleamine-pyrrole 2,3-dioxygenase (IDO) [20], T cell immunoreceptor with Immunoglobulin and ITIM domains (TIGIT) [21], CD96 [22] and many others [23].
LAG3 is the third inhibitory receptor pathway to be targeted in the clinic. LAG3 functions to control excessive activation following persistent antigen (Ag) exposure in an effort to prevent the onset of autoimmunity [24,25]; however, it can also contribute to a state of T cell dysfunction in the tumor microenvironment (TME) [24,26,27]. Dysfunctional T cells are characterized by impaired proliferation and cytokine production that distinguishes their inability to exert effector functions despite previous Ag encounters. Ineffective T cells have been detected in chronic inflammatory settings, including autoimmune diseases and tumors (i.e., tumor infiltrating lymphocytes or TIL). Various drugs targeting LAG3 are now available in the clinic with many more under development [28]. The aim of this review is to provide an overview of LAG3 biology under physiological and pathological conditions associated with chronic inflammatory microenvironments, such as that observed in various tumors including breast cancer (BC). Further, we include an overview of ongoing early phase clinical trials targeting LAG3 in oncology.

2. LAG3 Biological Activities and Expression

The LAG3 receptor is expressed on: 1) Activated human CD4+ (helper = Th) and CD8+ (cytotoxic = CTL) T cells where it is detectable within 24 h following in vitro stimulation [29]; 2) a subset of natural killer (NK) cells and invariant NK T cells [28,29]; and 3) murine plasmacytoid dendritic cells (pDC) where it was shown to be constitutively expressed [30], although this latter finding has not been confirmed. In addition, LAG3 expression was detected on B cells [31,32,33] and neurons [34], but these data also need validation. Apart from its membrane expression, LAG3 can also be stored in lysosomes, which facilitates its rapid appearance on the cell surface following T cell activation [35]. LAG3 principally interacts with major histocompatibility complex class II (MHC-II) molecules as its ligand, which is expressed on the surface of Ag presenting cells (APCs) and tumor cells [36,37] (Figure 1). A recent study has provided further insight by showing that LAG3+ CD4+ T cells selectively interact with stably expressed peptide-MHC-II complexes [38].
Activated effector CD4+ T cells, in particular regulatory T cells (Tregs) including both activated natural (nTregs) and induced CD4+ FoxP3+ Tregs (iTregs), express LAG3 [39] with their suppressive functions inhibited when it is blocked. Further, ectopic expression of LAG3 confers suppressive activity to non-Treg CD4+ T cells [38]. LAG3 inhibits CD4+ T cell activation via its conformational dependent recognition of stable peptide-MHC-II complexes [38]. A recent study found that LAG3 can also play a role in maintaining the quiescence of naïve CD4+ T cells [40]. LAG3 expression has been detected on highly immunosuppressive CD4+ FoxP3 IL10-secreting type 1 regulatory (Tr1) T cells, which have been identified in both humans and mice by dual expression of LAG3 and CD49b [41]. Tr1 cells are thought to play a role in promoting and maintaining tolerance in the periphery where they are induced. In cancer patients, LAG3 is preferentially expressed on Treg TIL in the TME relative to those in the periphery [41]. LAG3 can also inhibit APC (DC) functions by blocking their maturation when bound to LAG3+ Tregs [42].
LAG3 expression on CD8+ T and NK cells symbolizes a dysfunctional profile [43], although its effect on NK cells requires further investigation because blocking this pathway did not modify human NK cytotoxicity [43,44,45,46,47,48]. Because LAG3 impacts functionality, it has been suggested that it can bind ligands other than MHC-II, the latter constitutively expressed on APC and B cells, inducibly expressed on CD4+ T cells and other immune (neutrophils and eosinophils) and stromal cells (fibroblasts) and also expressed on many types of tumor cells [49,50] (Figure 1 and Figure 2).
Potential LAG3 ligands include: 1) Liver sinusoidal endothelial cell lectin (LSECtin), a member of the DC-SIGN family, which is expressed in the liver and on melanoma cells, where it functions to inhibit anti-tumor CD8+ T cell [51] and NK responses; 2) galectin-3, which is expressed by stromal cells [52,53] and CD8+ T cells in mice [54], which can bind LAG3 under conditions of heavy glycosylation [44]; 3) α-synuclein in neurons [55]; and the recently demonstrated fibrinogen-like protein 1, released in the liver (at low levels) and by tumor cells [34] ( Figure 1; Figure 2).
LAG3 also has a soluble form (sLAG3) released by shedding at the cell surface that provides an additional layer of control and immune regulation in the periphery or TME. sLAG3 is thought to impair monocyte differentiation into macrophages or DC, producing APC that ultimately have reduced immunostimulatory capacities [56]. It has also been studied as a circulating biomarker in BC patients with hormone receptor (HR)-positive metastatic disease, where detectable serum sLAG3 at diagnosis was associated with a survival advantage [57] with similar findings in a recent study of gastric cancer [58]. These data advocate further investigation of sLAG3 as a prognostic or predictive biomarker for LAG3-targeted and other cancer therapies.

3. LAG3 Expression at Sites Characterized by Chronic Immune Activation

LAG3 plays a protective role in autoimmune diseases by dampening CD4+ T cell responses through MHC-II engagement and inhibiting effector T cell responses by promoting Treg and Tr1 suppression. This checkpoint regulation, designed to curb excessive immune activities [59], was clearly demonstrated by its synergistic activity with PD-1 to prevent autoimmunity in mice [60]. In chronic infection models, LAG3 expression was directly correlated with infection severity [61]. Furthermore, it was co-expressed with PD-1 on dysfunctional virus-specific CD8+ T cells and parasite-specific CD4+ T cells [62]. Notably, while LAG3 blockade alone had little effect [61], the synergistic effect of adding PD-L1 blockade improved CD8+ [61] and CD4+ [62] T cell responses. The role LAG3 plays in the dysfunction of T cell TIL is similar to that observed during chronic viral infections. Following sustained Ag exposure activated CD4+ and CD8+ T cells upregulate multiple inhibitory receptors, and are characterized by impaired proliferation, cytokine secretion (IFNγ and TNFα) and cytolytic activity [59], as poorly functional lymphocytes.
LAG3 and PD-1 co-expression has been used to identify dysfunctional CD8+ T cells in human tumors with their co-blockade in ovarian cancer improving proliferation and cytokine production by tumor-Ag-specific CD8+ T cells [63]. Further, in metastatic ovarian cancer it was shown that LAG3 activity as a single agent was limited by the expression of other inhibitory immune checkpoint molecules (i.e. PD-1 and CTLA-4) [64]. In colorectal carcinoma, LAG3 was expressed at higher levels in microsatellite unstable compared with microsatellite stable tumors [65]. LAG3 was also shown to be highly expressed on Tregs in the peripheral blood, metastatic lymph nodes and tumor tissues from melanoma and colorectal carcinoma [66], head and neck squamous cell carcinoma and non-small cell lung cancer patients [67,68]. These LAG3+ Tregs have an activated, immunosuppressive phenotype and produce high levels of IL-10 and TGFβ1 [66].

4. LAG3 in Human Breast Cancer

A recent study by our group examined a series of targetable inhibitory immune checkpoint molecules in primary BC, finding that PD-1 and CTLA-4 were consistently highly expressed on specific T cell subpopulations [69]. We detected LAG3 expression on a small proportion of CD4+ and CD8+ TIL but only rarely on stromal and non-lymphoid cells in the TME. Remarkably, all tumors that expressed LAG3+ also contained PD-1+ CD4+ and/or CD8+ TIL. Generally, these cells expressed higher levels of PD-1 and, defined as PD-1 high (PD-1hi), which likely signals that these cells are either dysfunctional [69] or follicular helper T cells (Tfh) [70]. LAG3 gene expression by microarray data was associated with positive clinical outcomes in basal-like BC (~80% of triple negative BC) and was also frequently expressed with other immune checkpoint molecules such as TIM3. Several contemporaneous BC studies investigated LAG3 expression using immunohistochemistry (IHC) to correlate its presence with prognosis. In one study, intratumoral LAG3+ TIL were detected in 11% of tumors and significantly associated with more aggressive clinicopathological parameters, including: Young age, large tumor size, high proliferation, which also typically characterize the more aggressive HER2-enriched and basal-like BC subtypes [70]. Multivariate analyses identified BC patients with intratumoral LAG3+ TIL as having significantly improved BC specific survival (BCSS) (hazard ratio (HR): 0.71, 95% confidence interval (CI) 0.56–0.90), particularly if they were ER-negative (HR: 0.50, 95% CI 0.36–0.69). LAG3 on HER2-enriched and basal-like BC signifies that these patients have a good prognosis despite their diagnosis with an aggressive BC subtype. Interestingly, around half of PD-L1+ and two-thirds of PD-1+ tumors were also positive for intratumoral LAG3+ TIL in this study. Concurrent LAG3+ and CD8+ intratumoral TIL was also associated with increased BCSS (HR: 0.49, 95% CI 0.32–0.74). These data highlight the notion that not only LAG3 expression but the location of TIL expressing this checkpoint molecule can have a positive impact on clinical outcome in these aggressive BC subtypes.
PD-1 and LAG3 were shown to be concomitantly expressed in approximately 15% of TNBC patients and positively correlated with a TIL presence in another study [71]. However, PD-1 and LAG3 expression was not significantly associated with patient outcome in this dataset, potentially because authors did not perform spatial analysis of LAG3+ cells in the TME [72]. Their data identified an increased probability of achieving a pathologic complete response following neoadjuvant treatment if pre-treatment biopsies highly expressed LAG3, finding it was correlated with PD-1 and PD-L1 expression. Alternatively, in a recent study, patients with residual disease after neoadjuvant chemotherapy that were characterized by high LAG3 expression, and in particular associated with PD-L1 expression, had a poor prognosis [71]. Overall, these studies show that LAG3 expression in BC is associated with extensive immune infiltration and frequently associated with other immune checkpoint molecules (particularly PD-1/PD-L1), providing a strong rationale for targeting both LAG3 and PD-1/PD-L1 concurrently or sequentially. This idea is supported by the finding of LAG3 upregulation on TIL in MHC class II+ tumors that develop resistance to anti-PD-1 agents [73]. Some immune checkpoint molecules like TIGIT that are less expressed in breast tumors, which differs from CTLA-4, PD-1 and TIM3 [74] whose genes are upregulated, thus resulting more expressed in BC. On the contrary one study found that TIGIT was more frequently expressed than other the ligand for PD-1, PD-L1, in in situ lesions of the breast. Instead PD-L1 was more frequently detected in TN invasive tumors [75], signifying that the pattern of expression of these immune checkpoint molecules might vary in different phases of the disease (from in situ to invasive). In another study, peripheral blood from BC and colorectal cancer patients, observed LAG3 downregulation in patients with TIM3, TIGIT and PD-L1 upregulation [76], confirming the heterogeneous expression of these molecules also in circulating immune cells, and contributing to the complexity of this scenario.

5. LAG3 Blockade in Cancer and Clinical Trials Testing LAG3 Targeting Compounds

LAG3 regulates the proliferation, cytokine production and/or cytolytic functions of T cells through its cytoplasmic domain. Although most of the molecular mechanisms remain poorly understood (Figure 2), it is known that LAG3 recognition of stable peptide-MHC-II complexes is critical for activity [38]. Consequently, LAG3 expression signals that ongoing responses are active at the inflammatory site (i.e. the TME). The rationale for targeting LAG3 is based on the presence of a pre-existing immune response. Peptide-MHC-II complexes are generated following activation-induced by IFNγ release and LAG3 expression is dependent upon activation [77], providing the logic for clinically targeting LAG3. Blocking LAG3 has been shown to be active in association with anti-PD-1 or anti-PD-L1 agents in cancer patients. The main consequences of dual inhibition are: 1) Inhibiting Treg activities, 2) promoting DC maturation, and 3) rescuing dysfunctional CD4+ Th TIL and CD8+ CTL TIL (Figure 3).
The selection of LAG3 as partner with an anti-PD-1 or anti-PD-L1 blocking Ab has a hypothetical advantage with respect to CTLA-4 because of its lower toxicity. This however remains to be demonstrated in further studies and future clinical trials designed to compare these targets (i.e., CTLA-4 versus LAG3) as partners for anti-PD-1 or anti-PD-L1 agents. LAG3 is now being tested in clinical trials of patients previously treated with anti-PD-1/PD-L1 agents and preliminary data from melanoma patients has shown efficacy of this dual blockade [78]. Alternatively, CTLA-4 combined with the anti-PD-1 nivolumab given to immunotherapy naïve patients shows higher activity and efficacy coupled with a higher incidence rate of irAEs [79].
There are currently several agents targeting LAG3 that are being tested in clinical trials (summarized in Table 1). The earliest trial employed a soluble dimeric recombinant protein that contained four LAG3 extracellular domains in a soluble LAG3-immunoglobulin (Ig) named IMP321 [80]. This compound was initially designed to be a LAG3 antagonist, but it is currently being used as an immune adjuvant to activate APC because it binds with high affinity to MHC-II expressed on DC that mature and migrate to lymph nodes [81]. Early studies found that when the LAG3-Ig interacted with MHC-II on immature DC it induced CD80/86 upregulation, IL2 and TNFα secretion and promoted morphological changes in DC [82]. Its adjuvant function is to enhance cross presentation of Ag to T cells and thereby activate CD8+ T cell responses. IMP321, tested as a monotherapy (NCT00351949) or in combination with chemotherapy (NCT00732082 and NCT00349934) or vaccines [81], showed minimal activity in monotherapy [83] and modest activity using combination approaches [84].
Early data from the anti-LAG3 relatlimab used as simultaneous dual blockade targeting PD-1 together with LAG3 produced an objective response rate of 11% in melanoma patients progressing after anti-PD-1/PD-L1 monotherapy [85]. Translational data suggest that responses were associated with LAG3 expression in the TME, indicating that LAG3 might constitute a biomarker for patient selection. Relatlimab is currently being tested in microsatellite stable advanced colorectal carcinoma (NCT03642067), mismatch repair-deficient tumors resistant to prior PD-1/PD-L1 inhibition (NCT03607890), advanced chordoma (NCT03623854), advanced melanoma (NCT03470922, NCT02519322), gastro/esophageal cancer (NCT03044613, NCT03610711, NCT02935634) or gastroesophageal junction (NCT03662659), multiple solid cancers (NCT03459222, NCT01968109, NCT02966548, NCT03335540), advanced renal cell carcinoma (NCT02996110), virus-associated tumors (NCT02488759), non-small cell lung cancer (NCT02750514), hematological malignancies (NCT02061761) and glioblastoma (NCT03493932).
LAG3 antagonists have also been produced and are currently being tested in immunotherapy trials as a co-drug for simultaneously blocking the PD-1/PD-L1 pathway. The rationale for these trials is derived from preclinical data supporting its synergy with PD-1 blockade [86]. Bispecific Abs targeting LAG3 with Fc-domains for Ag binding to LAG3 were capable of binding human and murine LAG3, inhibiting its interaction with MHC-II and inducing IL-2 production in a T cell assay [28]. LAG3 is being targeted together with PD-1 in the dual immunomodulator MGD013 (ClinicalTrials.gov identifier: NCT03219268, phase I trial) whose objective is to inhibit both targets and induce a synergistic effect on tumor immunity, as previously shown in murine models [86]. Other drugs interacting with LAG3 include: LAG525, REGN3767, TSR-033 and GSK2831781 [87]. LAG525 is being tested for hematological malignancies (NCT03365791), TNBC (NCT03499899), advanced malignancies (NCT02460224) and metastatic melanoma (NCT03484923). REGN3767 is being investigated for advanced cancers (NCT03005782), TSR-033 in advanced solid tumors (NCT03250832) and GSK2831781 for psoriasis (NCT02195349).

6. Conclusions

The results from the various LAG3 clinical studies are currently unknown but the rationale behind them is based on data suggesting that co-targeting LAG3 is a promising approach for improving immunotherapy responses in multiple human tumor types. LAG3 co-expression with other immune checkpoint molecules, including PD-1, PD-L1 and TIM3, has been documented indicating that combinatorial immunotherapies targeting multiple TME immunosuppressive pathways hold promise. However, safety data must first demonstrate that simultaneous or sequential combinations are feasible and tolerable.

Author Contributions

C.S and K.W.G. did the bibliographic research, drafted the article and are responsible for data accuracy. P.D.S. and E.M. gave important intellectual input for writing the manuscript. C.S. created Figure 2; Figure 3. E.M. created Figure 1. K.W.G. supervised the entire work.

Funding

This research received no external funding.

Acknowledgments

Pushpamali De Silva is a fellow of the Belgian Fund for Scientific Research (FNRS)-Operation Télévie. We thank David Gray for assistance in writing in English.

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Khair, D.O.; Bax, H.J.; Mele, S.; Crescioli, S.; Pellizzari, G.; Khiabany, A.; Nakamura, M.; Harris, R.J.; French, E.; Hoffmann, R.M.; et al. Combining Immune Checkpoint Inhibitors: Established and Emerging Targets and Strategies to Improve Outcomes in Melanoma. Front. Immunol. 2019, 10, 453. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  2. Pacheco, J.M.; Camidge, D.R.; Doebele, R.C.; Schenk, E. A Changing of the Guard: Immune Checkpoint Inhibitors with and Without Chemotherapy as First Line Treatment for Metastatic Non-small Cell Lung Cancer. Front. Oncol. 2019, 9, 195. [Google Scholar] [CrossRef] [PubMed]
  3. Kammerer-Jacquet, S.F.; Deleuze, A.; Saout, J.; Mathieu, R.; Laguerre, B.; Verhoest, G.; Dugay, F.; Belaud-Rotureau, M.A.; Bensalah, K.; Rioux-Leclercq, N. Targeting the PD-1/PD-L1 Pathway in Renal Cell Carcinoma. Int. J. Mol. Sci. 2019, 20, 1692. [Google Scholar] [CrossRef] [PubMed]
  4. Von der Grun, J.; Rodel, F.; Brandts, C.; Fokas, E.; Guckenberger, M.; Rodel, C.; Balermpas, P. Targeted Therapies and Immune-Checkpoint Inhibition in Head and Neck Squamous Cell Carcinoma: Where Do We Stand Today and Where to Go? Cancers 2019, 11, 472. [Google Scholar] [CrossRef] [PubMed]
  5. Stahl, M.; Goldberg, A.D. Immune Checkpoint Inhibitors in Acute Myeloid Leukemia: Novel Combinations and Therapeutic Targets. Curr. Oncol. Rep. 2019, 21, 37. [Google Scholar] [CrossRef]
  6. Esteva, F.J.; Hubbard-Lucey, V.M.; Tang, J.; Pusztai, L. Immunotherapy and targeted therapy combinations in metastatic breast cancer. Lancet Oncol. 2019, 20, e175–e186. [Google Scholar] [CrossRef]
  7. Doroshow, D.B.; Sanmamed, M.F.; Hastings, K.; Politi, K.; Rimm, D.L.; Chen, L.; Melero, I.; Schalper, K.A.; Herbst, R.S. Immunotherapy in Non-Small Cell Lung Cancer: Facts and Hopes. Clin. Cancer Res. Off. J. Am. Assoc. Cancer Res. 2019. [Google Scholar] [CrossRef]
  8. Caccese, M.; Indraccolo, S.; Zagonel, V.; Lombardi, G. PD-1/PD-L1 immune-checkpoint inhibitors in glioblastoma: A concise review. Crit. Rev. Oncol. Hematol. 2019, 135, 128–134. [Google Scholar] [CrossRef]
  9. Lattanzi, M.; Balar, A.V. Current Status and Future Direction of Immunotherapy in Urothelial Carcinoma. Curr. Oncol. Rep. 2019, 21, 24. [Google Scholar] [CrossRef]
  10. Alhalabi, O.; Shah, A.Y.; Lemke, E.A.; Gao, J. Current and Future Landscape of Immune Checkpoint Inhibitors in Urothelial Cancer. Oncology 2019, 33, 11–18. [Google Scholar]
  11. Franssen, L.E.; Mutis, T.; Lokhorst, H.M.; van de Donk, N. Immunotherapy in myeloma: How far have we come? Ther. Adv. Hematol. 2019, 10, 2040620718822660. [Google Scholar] [CrossRef] [PubMed]
  12. Sandigursky, S.; Mor, A. Immune-Related Adverse Events in Cancer Patients Treated With Immune Checkpoint Inhibitors. Curr. Rheumatol. Rep. 2018, 20, 65. [Google Scholar] [CrossRef] [PubMed]
  13. Bhandari, S.; Gill, A.S.; Perez, C.A.; Jain, D. Management of immunotherapy toxicities in older adults. Semin. Oncol. 2018, 45, 226–231. [Google Scholar] [CrossRef] [PubMed]
  14. Porcu, M.; De Silva, P.; Solinas, C.; Battaglia, A.; Schena, M.; Scartozzi, M.; Bron, D.; Suri, J.S.; Willard-Gallo, K.; Sangiolo, D.; et al. Immunotherapy Associated Pulmonary Toxicity: Biology Behind Clinical and Radiological Features. Cancers 2019, 11, 305. [Google Scholar] [CrossRef] [PubMed]
  15. Kumar, P.; Saini, S.; Prabhakar, B.S. Cancer immunotherapy with check point inhibitor can cause autoimmune adverse events due to loss of Treg homeostasis. Semin. Cancer Biol. 2019. [Google Scholar] [CrossRef] [PubMed]
  16. Ruggeri, R.M.; Campenni, A.; Giuffrida, G.; Trimboli, P.; Giovanella, L.; Trimarchi, F.; Cannavo, S. Endocrine and metabolic adverse effects of immune checkpoint inhibitors: An overview (what endocrinologists should know). J. Endocrinol. Investig. 2018. [Google Scholar] [CrossRef] [PubMed]
  17. Solinas, C.; Porcu, M.; De Silva, P.; Musi, M.; Aspeslagh, S.; Scartozzi, M.; Willard-Gallo, K.; Mariotti, S.; Saba, L. Cancer immunotherapy-associated hypophysitis. Semin. Oncol. 2018, 45, 181–186. [Google Scholar] [CrossRef]
  18. Solinas, C.; De Silva, P.; Bron, D.; Willard-Gallo, K.D.S. Significance of TIM3 expression in cancer: From biology to the clinic. Under review.
  19. Sek, K.; Molck, C.; Stewart, G.D.; Kats, L.; Darcy, P.K.; Beavis, P.A. Targeting Adenosine Receptor Signaling in Cancer Immunotherapy. Int. J. Mol. Sci. 2018, 19, 3837. [Google Scholar] [CrossRef]
  20. Zhu, M.M.T.; Dancsok, A.R.; Nielsen, T.O. Indoleamine Dioxygenase Inhibitors: Clinical Rationale and Current Development. Curr. Oncol. Rep. 2019, 21, 2. [Google Scholar] [CrossRef]
  21. Solomon, B.L.; Garrido-Laguna, I. TIGIT: A novel immunotherapy target moving from bench to bedside. Cancer Immunol. Immunother. CII 2018, 67, 1659–1667. [Google Scholar] [CrossRef]
  22. Dougall, W.C.; Kurtulus, S.; Smyth, M.J.; Anderson, A.C. TIGIT and CD96: New checkpoint receptor targets for cancer immunotherapy. Immunol. Rev. 2017, 276, 112–120. [Google Scholar] [CrossRef] [PubMed]
  23. Hahn, A.W.; Gill, D.M.; Pal, S.K.; Agarwal, N. The future of immune checkpoint cancer therapy after PD-1 and CTLA-4. Immunotherapy 2017, 9, 681–692. [Google Scholar] [CrossRef] [PubMed]
  24. Zhang, Q.; Chikina, M.; Szymczak-Workman, A.L.; Horne, W.; Kolls, J.K.; Vignali, K.M.; Normolle, D.; Bettini, M.; Workman, C.J.; Vignali, D.A.A. LAG3 limits regulatory T cell proliferation and function in autoimmune diabetes. Sci. Immunol. 2017, 2. [Google Scholar] [CrossRef] [PubMed]
  25. Kadowaki, A.; Miyake, S.; Saga, R.; Chiba, A.; Mochizuki, H.; Yamamura, T. Gut environment-induced intraepithelial autoreactive CD4(+) T cells suppress central nervous system autoimmunity via LAG-3. Nat. Commun. 2016, 7, 11639. [Google Scholar] [CrossRef] [PubMed]
  26. Van der Zwan, A.; Bi, K.; Norwitz, E.R.; Crespo, A.C.; Claas, F.H.J.; Strominger, J.L.; Tilburgs, T. Mixed signature of activation and dysfunction allows human decidual CD8(+) T cells to provide both tolerance and immunity. Proc. Natl. Acad. Sci. USA 2018, 115, 385–390. [Google Scholar] [CrossRef]
  27. Gameiro, S.F.; Ghasemi, F.; Barrett, J.W.; Koropatnick, J.; Nichols, A.C.; Mymryk, J.S.; Maleki Vareki, S. Treatment-naive HPV+ head and neck cancers display a T-cell-inflamed phenotype distinct from their HPV- counterparts that has implications for immunotherapy. Oncoimmunology 2018, 7, e1498439. [Google Scholar] [CrossRef] [PubMed]
  28. Everett, K.L.; Kraman, M.; Wollerton, F.P.G.; Zimarino, C.; Kmiecik, K.; Gaspar, M.; Pechouckova, S.; Allen, N.L.; Doody, J.F.; Tuna, M. Generation of Fcabs targeting human and murine LAG-3 as building blocks for novel bispecific antibody therapeutics. Methods 2018. [Google Scholar] [CrossRef]
  29. Workman, C.J.; Rice, D.S.; Dugger, K.J.; Kurschner, C.; Vignali, D.A. Phenotypic analysis of the murine CD4-related glycoprotein, CD223 (LAG-3). Eur. J. Immunol. 2002, 32, 2255–2263. [Google Scholar] [CrossRef]
  30. Workman, C.J.; Wang, Y.; El Kasmi, K.C.; Pardoll, D.M.; Murray, P.J.; Drake, C.G.; Vignali, D.A. LAG-3 regulates plasmacytoid dendritic cell homeostasis. J. Immunol. 2009, 182, 1885–1891. [Google Scholar] [CrossRef]
  31. Kisielow, M.; Kisielow, J.; Capoferri-Sollami, G.; Karjalainen, K. Expression of lymphocyte activation gene 3 (LAG-3) on B cells is induced by T cells. Eur. J. Immunol. 2005, 35, 2081–2088. [Google Scholar] [CrossRef]
  32. Kotaskova, J.; Tichy, B.; Trbusek, M.; Francova, H.S.; Kabathova, J.; Malcikova, J.; Doubek, M.; Brychtova, Y.; Mayer, J.; Pospisilova, S. High expression of lymphocyte-activation gene 3 (LAG3) in chronic lymphocytic leukemia cells is associated with unmutated immunoglobulin variable heavy chain region (IGHV) gene and reduced treatment-free survival. J. Mol. Diagn. Jmd 2010, 12, 328–334. [Google Scholar] [CrossRef] [PubMed]
  33. Lino, A.C.; Dang, V.D.; Lampropoulou, V.; Welle, A.; Joedicke, J.; Pohar, J.; Simon, Q.; Thalmensi, J.; Baures, A.; Fluhler, V.; et al. LAG-3 Inhibitory Receptor Expression Identifies Immunosuppressive Natural Regulatory Plasma Cells. Immunity 2018, 49, 120–133 e9. [Google Scholar] [CrossRef] [PubMed]
  34. Mao, X.; Ou, M.T.; Karuppagounder, S.S.; Kam, T.I.; Yin, X.; Xiong, Y.; Ge, P.; Umanah, G.E.; Brahmachari, S.; Shin, J.H.; et al. Pathological alpha-synuclein transmission initiated by binding lymphocyte-activation gene 3. Science 2016, 353. [Google Scholar] [CrossRef] [PubMed]
  35. Bae, J.; Lee, S.J.; Park, C.G.; Lee, Y.S.; Chun, T. Trafficking of LAG-3 to the surface on activated T cells via its cytoplasmic domain and protein kinase C signaling. J. Immunol. 2014, 193, 3101–3112. [Google Scholar] [CrossRef]
  36. Salgado, R.; Denkert, C.; Demaria, S.; Sirtaine, N.; Klauschen, F.; Pruneri, G.; Wienert, S.; Eynden, G.V.d.; Baehner, F.L.; Penault-Llorca, F.; et al. The evaluation of tumor-infiltrating lymphocytes (TILs) in breast cancer: Recommendations by an International TILs Working Group 2014. Ann. Oncol. 2014, 26, 259–271. [Google Scholar] [CrossRef] [PubMed]
  37. Baixeras, E.; Huard, B.; Miossec, C.; Jitsukawa, S.; Martin, M.; Hercend, T.; Auffray, C.; Triebel, F.; Piatier-Tonneau, D. Characterization of the lymphocyte activation gene 3-encoded protein. A new ligand for human leukocyte antigen class II antigens. J. Exp. Med. 1992, 176, 327–337. [Google Scholar] [CrossRef]
  38. Maruhashi, T.; Okazaki, I.M.; Sugiura, D.; Takahashi, S.; Maeda, T.K.; Shimizu, K.; Okazaki, T. LAG-3 inhibits the activation of CD4(+) T cells that recognize stable pMHCII through its conformation-dependent recognition of pMHCII. Nat. Immunol. 2018, 19, 1415–1426. [Google Scholar] [CrossRef] [PubMed]
  39. Huang, C.T.; Workman, C.J.; Flies, D.; Pan, X.; Marson, A.L.; Zhou, G.; Hipkiss, E.L.; Ravi, S.; Kowalski, J.; Levitsky, H.I.; et al. Role of LAG-3 in regulatory T cells. Immunity 2004, 21, 503–513. [Google Scholar] [CrossRef]
  40. Previte, D.M.; Martins, C.P.; O’Connor, E.C.; Marre, M.L.; Coudriet, G.M.; Beck, N.W.; Menk, A.V.; Wright, R.H.; Tse, H.M.; Delgoffe, G.M.; et al. Lymphocyte Activation Gene-3 Maintains Mitochondrial and Metabolic Quiescence in Naive CD4(+) T Cells. Cell Rep. 2019, 27, 129–141 e4. [Google Scholar] [CrossRef]
  41. Gagliani, N.; Magnani, C.F.; Huber, S.; Gianolini, M.E.; Pala, M.; Licona-Limon, P.; Guo, B.; Herbert, D.R.; Bulfone, A.; Trentini, F.; et al. Coexpression of CD49b and LAG-3 identifies human and mouse T regulatory type 1 cells. Nat. Med. 2013, 19, 739–746. [Google Scholar] [CrossRef]
  42. Liang, B.; Workman, C.; Lee, J.; Chew, C.; Dale, B.M.; Colonna, L.; Flores, M.; Li, N.; Schweighoffer, E.; Greenberg, S.; et al. Regulatory T cells inhibit dendritic cells by lymphocyte activation gene-3 engagement of MHC class II. J. Immunol. 2008, 180, 5916–5926. [Google Scholar] [CrossRef] [PubMed]
  43. Huard, B.; Tournier, M.; Triebel, F. LAG-3 does not define a specific mode of natural killing in human. Immunol. Lett. 1998, 61, 109–112. [Google Scholar] [CrossRef]
  44. Graydon, C.G.; Balasko, A.L.; Fowke, K.R. Roles, function and relevance of LAG3 in HIV infection. PLoS Pathog. 2019, 15, e1007429. [Google Scholar] [CrossRef] [PubMed]
  45. Kurachi, M. CD8(+) T cell exhaustion. Semin. Immunopathol. 2019, 41, 327–337. [Google Scholar] [CrossRef] [PubMed]
  46. McLane, L.M.; Abdel-Hakeem, M.S.; Wherry, E.J. CD8 T Cell Exhaustion During Chronic Viral Infection and Cancer. Annu. Rev. Immunol. 2019, 37, 457–495. [Google Scholar] [CrossRef] [PubMed]
  47. Edwards, J.; Wilmott, J.S.; Madore, J.; Gide, T.N.; Quek, C.; Tasker, A.; Ferguson, A.; Chen, J.; Hewavisenti, R.; Hersey, P.; et al. CD103(+) Tumor-Resident CD8(+) T Cells Are Associated with Improved Survival in Immunotherapy-Naive Melanoma Patients and Expand Significantly During Anti-PD-1 Treatment. Clin. Cancer Res. Off. J. Am. Assoc. Cancer Res. 2018, 24, 3036–3045. [Google Scholar] [CrossRef] [PubMed]
  48. Martinet, L.; Smyth, M.J. Balancing natural killer cell activation through paired receptors. Nat. Rev. Immunol. 2015, 15, 243–254. [Google Scholar] [CrossRef] [PubMed]
  49. Jurewicz, M.M.; Stern, L.J. Class II MHC antigen processing in immune tolerance and inflammation. Immunogenetics 2019, 71, 171–187. [Google Scholar] [CrossRef] [PubMed]
  50. Axelrod, M.L.; Cook, R.S.; Johnson, D.B.; Balko, J.M. Biological Consequences of MHC-II Expression by Tumor Cells in Cancer. Clin. Cancer Res. Off. J. Am. Assoc. Cancer Res. 2019, 25, 2392–2402. [Google Scholar] [CrossRef] [PubMed]
  51. Xu, F.; Liu, J.; Liu, D.; Liu, B.; Wang, M.; Hu, Z.; Du, X.; Tang, L.; He, F. LSECtin expressed on melanoma cells promotes tumor progression by inhibiting antitumor T-cell responses. Cancer Res. 2014, 74, 3418–3428. [Google Scholar] [CrossRef] [PubMed]
  52. Fei, F.; Joo, E.J.; Tarighat, S.S.; Schiffer, I.; Paz, H.; Fabbri, M.; Abdel-Azim, H.; Groffen, J.; Heisterkamp, N. B-cell precursor acute lymphoblastic leukemia and stromal cells communicate through Galectin-3. Oncotarget 2015, 6, 11378–11394. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  53. Ruvolo, P.P. Galectin 3 as a guardian of the tumor microenvironment. Biochim. Et Biophys. Acta 2016, 1863, 427–437. [Google Scholar] [CrossRef] [PubMed]
  54. Kaur, M.; Kumar, D.; Butty, V.; Singh, S.; Esteban, A.; Fink, G.R.; Ploegh, H.L.; Sehrawat, S. Galectin-3 Regulates gamma-Herpesvirus Specific CD8 T Cell Immunity. iScience 2018, 9, 101–119. [Google Scholar] [CrossRef] [PubMed]
  55. Wang, J.; Sanmamed, M.F.; Datar, I.; Su, T.T.; Ji, L.; Sun, J.; Chen, L.; Chen, Y.; Zhu, G.; Yin, W.; et al. Fibrinogen-like Protein 1 Is a Major Immune Inhibitory Ligand of LAG-3. Cell 2019, 176, 334–347. [Google Scholar] [CrossRef] [PubMed]
  56. Lienhardt, C.; Azzurri, A.; Amedei, A.; Fielding, K.; Sillah, J.; Sow, O.Y.; Bah, B.; Benagiano, M.; Diallo, A.; Manetti, R.; et al. Active tuberculosis in Africa is associated with reduced Th1 and increased Th2 activity in vivo. Eur. J. Immunol. 2002, 32, 1605–1613. [Google Scholar] [CrossRef] [Green Version]
  57. Triebel, F.; Hacene, K.; Pichon, M.F. A soluble lymphocyte activation gene-3 (sLAG-3) protein as a prognostic factor in human breast cancer expressing estrogen or progesterone receptors. Cancer Lett. 2006, 235, 147–153. [Google Scholar] [CrossRef] [PubMed]
  58. Li, N.; Jilisihan, B.; Wang, W.; Tang, Y.; Keyoumu, S. Soluble LAG3 acts as a potential prognostic marker of gastric cancer and its positive correlation with CD8+T cell frequency and secretion of IL-12 and INF-gamma in peripheral blood. Cancer Biomark. Sect. A Dis. Markers 2018, 23, 341–351. [Google Scholar] [CrossRef]
  59. Anderson, A.C.; Joller, N.; Kuchroo, V.K. Lag-3, Tim-3, and TIGIT: Co-inhibitory Receptors with Specialized Functions in Immune Regulation. Immunity 2016, 44, 989–1004. [Google Scholar] [CrossRef] [Green Version]
  60. Okazaki, T.; Okazaki, I.M.; Wang, J.; Sugiura, D.; Nakaki, F.; Yoshida, T.; Kato, Y.; Fagarasan, S.; Muramatsu, M.; Eto, T.; et al. PD-1 and LAG-3 inhibitory co-receptors act synergistically to prevent autoimmunity in mice. J. Exp. Med. 2011, 208, 395–407. [Google Scholar] [CrossRef] [Green Version]
  61. Blackburn, S.D.; Shin, H.; Haining, W.N.; Zou, T.; Workman, C.J.; Polley, A.; Betts, M.R.; Freeman, G.J.; Vignali, D.A.; Wherry, E.J. Coregulation of CD8+ T cell exhaustion by multiple inhibitory receptors during chronic viral infection. Nat. Immunol. 2009, 10, 29–37. [Google Scholar] [CrossRef]
  62. Butler, N.S.; Moebius, J.; Pewe, L.L.; Traore, B.; Doumbo, O.K.; Tygrett, L.T.; Waldschmidt, T.J.; Crompton, P.D.; Harty, J.T. Therapeutic blockade of PD-L1 and LAG-3 rapidly clears established blood-stage Plasmodium infection. Nat. Immunol. 2011, 13, 188–195. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  63. Matsuzaki, J.; Gnjatic, S.; Mhawech-Fauceglia, P.; Beck, A.; Miller, A.; Tsuji, T.; Eppolito, C.; Qian, F.; Lele, S.; Shrikant, P.; et al. Tumor-infiltrating NY-ESO-1-specific CD8+ T cells are negatively regulated by LAG-3 and PD-1 in human ovarian cancer. Proc. Natl. Acad. Sci. USA 2010, 107, 7875–7880. [Google Scholar] [CrossRef] [PubMed]
  64. Huang, R.Y.; Francois, A.; McGray, A.R.; Miliotto, A.; Odunsi, K. Compensatory upregulation of PD-1, LAG-3, and CTLA-4 limits the efficacy of single-agent checkpoint blockade in metastatic ovarian cancer. Oncoimmunology 2017, 6, e1249561. [Google Scholar] [CrossRef] [PubMed]
  65. Llosa, N.J.; Cruise, M.; Tam, A.; Wicks, E.C.; Hechenbleikner, E.M.; Taube, J.M.; Blosser, R.L.; Fan, H.; Wang, H.; Luber, B.S.; et al. The vigorous immune microenvironment of microsatellite instable colon cancer is balanced by multiple counter-inhibitory checkpoints. Cancer Discov. 2015, 5, 43–51. [Google Scholar] [CrossRef] [PubMed]
  66. Camisaschi, C.; Casati, C.; Rini, F.; Perego, M.; De Filippo, A.; Triebel, F.; Parmiani, G.; Belli, F.; Rivoltini, L.; Castelli, C. LAG-3 expression defines a subset of CD4(+)CD25(high)Foxp3(+) regulatory T cells that are expanded at tumor sites. J. Immunol. 2010, 184, 6545–6551. [Google Scholar] [CrossRef] [PubMed]
  67. Jie, H.B.; Gildener-Leapman, N.; Li, J.; Srivastava, R.M.; Gibson, S.P.; Whiteside, T.L.; Ferris, R.L. Intratumoral regulatory T cells upregulate immunosuppressive molecules in head and neck cancer patients. Br. J. Cancer 2013, 109, 2629–2635. [Google Scholar] [CrossRef] [PubMed]
  68. Wei, T.; Zhang, J.; Qin, Y.; Wu, Y.; Zhu, L.; Lu, L.; Tang, G.; Shen, Q. Increased expression of immunosuppressive molecules on intratumoral and circulating regulatory T cells in non-small-cell lung cancer patients. Am. J. Cancer Res. 2015, 5, 2190–2201. [Google Scholar] [PubMed]
  69. Solinas, C.; Garaud, S.; De Silva, P.; Boisson, A.; Van den Eynden, G.; de Wind, A.; Risso, P.; Rodrigues Vitória, J.; Richard, F.; Migliori, E.; et al. Immune Checkpoint Molecules on Tumor-Infiltrating Lymphocytes and Their Association with Tertiary Lymphoid Structures in Human Breast Cancer. Front. Immunol. 2017, 7. [Google Scholar] [CrossRef]
  70. Burugu, S.; Gao, D.; Leung, S.; Chia, S.K.; Nielsen, T.O. LAG-3+ tumor infiltrating lymphocytes in breast cancer: Clinical correlates and association with PD-1/PD-L1+ tumors. Ann. Oncol. Off. J. Eur. Soc. Med Oncol. 2017, 28, 2977–2984. [Google Scholar] [CrossRef]
  71. Wang, Y.; Dong, T.; Xuan, Q.; Zhao, H.; Qin, L.; Zhang, Q. Lymphocyte-Activation Gene-3 Expression and Prognostic Value in Neoadjuvant-Treated Triple-Negative Breast Cancer. J. Breast Cancer 2018, 21, 124–133. [Google Scholar] [CrossRef]
  72. Bottai, G.; Raschioni, C.; Losurdo, A.; Di Tommaso, L.; Tinterri, C.; Torrisi, R.; Reis-Filho, J.S.; Roncalli, M.; Sotiriou, C.; Santoro, A.; et al. An immune stratification reveals a subset of PD-1/LAG-3 double-positive triple-negative breast cancers. Breast Cancer Res. BCR 2016, 18, 121. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  73. Johnson, D.B.; Nixon, M.J.; Wang, Y.; Wang, D.Y.; Castellanos, E.; Estrada, M.V.; Ericsson-Gonzalez, P.I.; Cote, C.H.; Salgado, R.; Sanchez, V.; et al. Tumor-specific MHC-II expression drives a unique pattern of resistance to immunotherapy via LAG-3/FCRL6 engagement. JCI Insight 2018, 3. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  74. Sasidharan Nair, V.; El Salhat, H.; Taha, R.Z.; John, A.; Ali, B.R.; Elkord, E. DNA methylation and repressive H3K9 and H3K27 trimethylation in the promoter regions of PD-1, CTLA-4, TIM-3, LAG-3, TIGIT, and PD-L1 genes in human primary breast cancer. Clin. Epigenetics 2018, 10, 78. [Google Scholar] [CrossRef] [PubMed]
  75. Gil Del Alcazar, C.R.; Huh, S.J.; Ekram, M.B.; Trinh, A.; Liu, L.L.; Beca, F.; Zi, X.; Kwak, M.; Bergholtz, H.; Su, Y.; et al. Immune Escape in Breast Cancer During In Situ to Invasive Carcinoma Transition. Cancer Discov. 2017, 7, 1098–1115. [Google Scholar] [CrossRef] [PubMed]
  76. Elashi, A.A.; Sasidharan Nair, V.; Taha, R.Z.; Shaath, H.; Elkord, E. DNA methylation of immune checkpoints in the peripheral blood of breast and colorectal cancer patients. Oncoimmunology 2019, 8, e1542918. [Google Scholar] [CrossRef]
  77. Lui, Y.; Davis, S.J. LAG-3: A very singular immune checkpoint. Nat. Immunol. 2018, 19, 1278–1279. [Google Scholar] [CrossRef] [PubMed]
  78. Ascierto, P.A.; Melero, I.; Bhatia, S.; Bono, P.; Sanborn, R.E.; Lipson, E.J.; Callahan, M.K.; Gajewski, T.; Gomez-Roca, C.A.; Hodi, F.S.; et al. Initial efficacy of anti-lymphocyte activation gene-3 (anti–LAG-3; BMS-986016) in combination with nivolumab (nivo) in pts with melanoma (MEL) previously treated with anti–PD-1/PD-L1 therapy. J. Clin. Oncol. 2017, 35, 9520. [Google Scholar] [CrossRef]
  79. Hodi, F.S.; Chiarion-Sileni, V.; Gonzalez, R.; Grob, J.J.; Rutkowski, P.; Cowey, C.L.; Lao, C.D.; Schadendorf, D.; Wagstaff, J.; Dummer, R.; et al. Nivolumab plus ipilimumab or nivolumab alone versus ipilimumab alone in advanced melanoma (CheckMate 067): 4-year outcomes of a multicentre, randomised, phase 3 trial. Lancet Oncol. 2018, 19, 1480–1492. [Google Scholar] [CrossRef]
  80. Prigent, P.; El Mir, S.; Dreano, M.; Triebel, F. Lymphocyte activation gene-3 induces tumor regression and antitumor immune responses. Eur. J. Immunol. 1999, 29, 3867–3876. [Google Scholar] [CrossRef]
  81. Romano, E.; Michielin, O.; Voelter, V.; Laurent, J.; Bichat, H.; Stravodimou, A.; Romero, P.; Speiser, D.E.; Triebel, F.; Leyvraz, S.; et al. MART-1 peptide vaccination plus IMP321 (LAG-3Ig fusion protein) in patients receiving autologous PBMCs after lymphodepletion: Results of a Phase I trial. J. Transl. Med. 2014, 12, 97. [Google Scholar] [CrossRef]
  82. Avice, M.N.; Sarfati, M.; Triebel, F.; Delespesse, G.; Demeure, C.E. Lymphocyte activation gene-3, a MHC class II ligand expressed on activated T cells, stimulates TNF-alpha and IL-12 production by monocytes and dendritic cells. J. Immunol. 1999, 162, 2748–2753. [Google Scholar] [PubMed]
  83. Brignone, C.; Escudier, B.; Grygar, C.; Marcu, M.; Triebel, F. A phase I pharmacokinetic and biological correlative study of IMP321, a novel MHC class II agonist, in patients with advanced renal cell carcinoma. Clin. Cancer Res. Off. J. Am. Assoc. Cancer Res. 2009, 15, 6225–6231. [Google Scholar] [CrossRef] [PubMed]
  84. Wang-Gillam, A.; Plambeck-Suess, S.; Goedegebuure, P.; Simon, P.O.; Mitchem, J.B.; Hornick, J.R.; Sorscher, S.; Picus, J.; Suresh, R.; Lockhart, A.C.; et al. A phase I study of IMP321 and gemcitabine as the front-line therapy in patients with advanced pancreatic adenocarcinoma. Investig. New Drugs 2013, 31, 707–713. [Google Scholar] [CrossRef] [PubMed]
  85. Ascierto, P.A.; Bono, P.; Bhatia, S.; Melero, I.; Nyakas, M.S.; Svane, I.-M.; Larkin, J.; Gomez-Roca, C.; Schadendorf, D.; Dummer, R.; et al. LBA18. Efficacy of BMS-986016, a monoclonal antibody that targets lymphocyte activation gene-3 (LAG-3), in combination with nivolumab in pts with melanoma who progressed during prior anti–PD-1/PD-L1 therapy (mel prior IO) in all-comer and biomarker-enriched populations. Ann. Oncol. 2017, 28, mdx440.011. [Google Scholar] [CrossRef]
  86. Woo, S.R.; Turnis, M.E.; Goldberg, M.V.; Bankoti, J.; Selby, M.; Nirschl, C.J.; Bettini, M.L.; Gravano, D.M.; Vogel, P.; Liu, C.L.; et al. Immune inhibitory molecules LAG-3 and PD-1 synergistically regulate T-cell function to promote tumoral immune escape. Cancer Res. 2012, 72, 917–927. [Google Scholar] [CrossRef] [PubMed]
  87. Ko, E.C.; Raben, D.; Formenti, S.C. The Integration of Radiotherapy with Immunotherapy for the Treatment of Non-Small Cell Lung Cancer. Clin. Cancer Res. Off. J. Am. Assoc. Cancer Res. 2018. [Google Scholar] [CrossRef] [PubMed]
Figure 1. LAG3 biology on immune cells (A) and in the tumor microenvironment (TME) (B). (A). LAG3 is expressed on CD4+ (Th) and CD8+ (CTL) T cells, plasmacytoid dendritic cells (pDC) and NK cells in the TME (B). Its principle ligands include: MHC-II expressed on antigen presenting cells (APC) and tumor cells, LSECtin expressed on melanoma cells and galectin-3 expressed on some T cells and stromal cells in the TME (B). In its soluble form, LAG3 (sLAG3) impairs monocyte differentiation to dendritic cells (DC) and macrophages. In the TME, interactions mediated by LAG3 and its ligands are inhibitory (B).
Figure 1. LAG3 biology on immune cells (A) and in the tumor microenvironment (TME) (B). (A). LAG3 is expressed on CD4+ (Th) and CD8+ (CTL) T cells, plasmacytoid dendritic cells (pDC) and NK cells in the TME (B). Its principle ligands include: MHC-II expressed on antigen presenting cells (APC) and tumor cells, LSECtin expressed on melanoma cells and galectin-3 expressed on some T cells and stromal cells in the TME (B). In its soluble form, LAG3 (sLAG3) impairs monocyte differentiation to dendritic cells (DC) and macrophages. In the TME, interactions mediated by LAG3 and its ligands are inhibitory (B).
Cancers 11 01213 g001
Figure 2. Molecular mechanisms of LAG3 function. LAG3 can be expressed on CD4+ (A), CD8+ T and NK cells (B). Its interaction with its ligands (stable pMHC-II complexes; LSECtin; FGL-1; galectin-3) expressed on different cells (immune, stromal, liver and tumor cells), generates an inhibition of CD4+, CD8+ and NK T cell proliferation, cytokine production and cytolitic function. These effects are mediated by the cytoplasmic motif of the LAG3 receptor, which is named KIEELE.
Figure 2. Molecular mechanisms of LAG3 function. LAG3 can be expressed on CD4+ (A), CD8+ T and NK cells (B). Its interaction with its ligands (stable pMHC-II complexes; LSECtin; FGL-1; galectin-3) expressed on different cells (immune, stromal, liver and tumor cells), generates an inhibition of CD4+, CD8+ and NK T cell proliferation, cytokine production and cytolitic function. These effects are mediated by the cytoplasmic motif of the LAG3 receptor, which is named KIEELE.
Cancers 11 01213 g002
Figure 3. Targeting effector and regulatory T cells with LAG3 antagonistic antibodies (A) and activating antigen presenting cells with soluble LAG3 Immunoglobulin (Ig) (B). (A) Binding of LAG3 antagonistic antibodies (Abs) to LAG3 prevents its interaction with the respective ligands (LSECtin, galectin-3 and FGL-1 for CD8+ T cells; antigen presenting cells (APC) for CD4+ T cells). This contributes to releasing the break to the activation of tumor-infiltrating lymphocytes (TIL) and is achieved with the use of a combined blockade with anti-PD-1/PD-L1 Abs. (B) Binding of a soluble LAG3 Ig to APC engages MHC-II molecules on the cell surface. This generates an upregulation of co-stimulatory molecules, such as CD80, CD86 and CD40, and leads to the secretion of pro-inflammatory cytokines and chemokines by APC. APC activation through soluble LAG3 agonistic Ab may induce a rapid release of pro-inflammatory cytokines by a subpopulation of NK cells and activation of memory CD8+ T cells in an antigen independent manner. APC: Antigen presenting cell; CCL: chemokine (C-C motif) ligand; CXCL: chemokine (C-X-C motif) ligand; DC: dendritic cell; FGL-1: fibrinoigen like protein - 1; IFN: interferon; Ig: immunoglobulin; IL: interleukin; LSECtin: liver and lymph node sinusoidal endothelial cell C-type lectin; NK: natural killer; PD-1: programmed cell death -1; Teff: effector T cells; TNF: tumor necrosis factor; Tregs: regulatory T cells.
Figure 3. Targeting effector and regulatory T cells with LAG3 antagonistic antibodies (A) and activating antigen presenting cells with soluble LAG3 Immunoglobulin (Ig) (B). (A) Binding of LAG3 antagonistic antibodies (Abs) to LAG3 prevents its interaction with the respective ligands (LSECtin, galectin-3 and FGL-1 for CD8+ T cells; antigen presenting cells (APC) for CD4+ T cells). This contributes to releasing the break to the activation of tumor-infiltrating lymphocytes (TIL) and is achieved with the use of a combined blockade with anti-PD-1/PD-L1 Abs. (B) Binding of a soluble LAG3 Ig to APC engages MHC-II molecules on the cell surface. This generates an upregulation of co-stimulatory molecules, such as CD80, CD86 and CD40, and leads to the secretion of pro-inflammatory cytokines and chemokines by APC. APC activation through soluble LAG3 agonistic Ab may induce a rapid release of pro-inflammatory cytokines by a subpopulation of NK cells and activation of memory CD8+ T cells in an antigen independent manner. APC: Antigen presenting cell; CCL: chemokine (C-C motif) ligand; CXCL: chemokine (C-X-C motif) ligand; DC: dendritic cell; FGL-1: fibrinoigen like protein - 1; IFN: interferon; Ig: immunoglobulin; IL: interleukin; LSECtin: liver and lymph node sinusoidal endothelial cell C-type lectin; NK: natural killer; PD-1: programmed cell death -1; Teff: effector T cells; TNF: tumor necrosis factor; Tregs: regulatory T cells.
Cancers 11 01213 g003
Table 1. Ongoing clinical trials using LAG3 blockade to treat cancer patients.
Table 1. Ongoing clinical trials using LAG3 blockade to treat cancer patients.
ReferenceDrug(s)PhaseTumor TypeMain ObjectivesStatus
NCT
02676869
IMP321 (LAG3 Ig fusion protein-eftilagimod alpha) + pembrolizumab (anti-PD-1)IStage IV and stage III melanomaSafety, tolerability and recommended phase 2 doseActive, not yet recruiting
NCT
02614833
IMP321 (LAG3 Ig fusion protein-eftilagimod alpha) + paclitaxelIIb, randomizedHormone receptor-positive stage IV breast cancerRecommended phase II dose, safety and efficacy (survival and objective response rate)Recruiting
NCT
03252938
IMP321 (LAG3 Ig fusion protein-eftilagimod alpha)IMetastatic solid tumors; peritoneal carcinomatosisFeasibility and safety of intra-tumoral and intra-peritoneal (new routes of administration), and subcutaneous injectionsRecruiting
NCT
03625323
IMP321 (LAG3 Ig fusion protein-eftilagimod alpha) + pembrolizumab (anti-PD-1)IIAdvanced or metastatic non-small cell lung carcinoma and head and neck squamous cell carcinomaSafety and efficacy (objective response rate)Recruiting
NCT
03642067
Relatlimab (anti-LAG3 monoclonal antibody BMS-986016) + nivolumab (anti-PD-1)IIMicrosatellite stable (MSS) colorectal carcinomas
Colorectal carcinoma
Response (objective response rate) and biomarkersRecruiting
NCT
03623854
Relatlimab (anti-LAG3 monoclonal antibody BMS-986016) + nivolumab (anti-PD-1)IIAdvanced chordomasClinical benefits (objective response rate, progression free survival) and safetyNot yet recruiting
NCT
03743766
Relatlimab (anti-LAG3 monoclonal antibody BMS-986016) +/- nivolumab (anti-PD-1)IIStage III or stage IV melanomaChange in LAG3 and PD-1 expression; change in tumor size and overall response rateRecruiting
NCT
03607890
Relatlimab (anti-LAG3 monoclonal antibody BMS-986016) + nivolumab (anti-PD-1)IIRefractory microsatellite unstable high (MSI-H) solid tumors prior of PD-(L) 1 therapy
MSI-H tumors
Objective response rate, toxicity, survival, disease control rate, best overall response, duration of response, duration of clinical benefit, time to objective responseRecruiting
NCT
03724968
Relatlimab (anti-LAG3 monoclonal antibody BMS-986016) +/- nivolumab (anti-PD-1); nivolumab + ipilimumab (anti-CTLA-4)IIStage III and stage IV melanoma, stratified by MHC-II expressionEfficacy, measured by change in activated GZMB+ CD8+ T-cell density intratumorally; response rate, median progression free survival, overall survival, and safety and tolerability of nivolumab plus relatlimab in patients with MHC-II (+) melanoma, and of nivolumab plus ipilimumab in patients with MHC-II (-) melanoma; explore potential associations of biomarkers with clinical efficacy and/or incidence of adverse events due to study drug by analyzing biomarker measures within the peripheral blood and tumor microenvironmentRecruiting
NCT
03470922
Nivolumab (anti-PD-1) +/- relatlimab (anti-LAG3 monoclonal antibody BMS-986016)II/IIIPreviously untreated stage III or stage IV melanomaSurvival (progression free survival, overall survival), responses (objective response rate), duration of response, toxicityActive, not yet recruiting
NCT
03044613
Nivolumab (anti-PD-1) or Nivolumab plus relatlimab (anti-LAG3 monoclonal antibody BMS-986016); chemoradiationIbStage II/III gastric cancer, esophageal cancer, gastroesophageal cancerToxicity, feasibility, pathologic complete response rate; approximate quantitation of infused nivolumab bound to PD-1 receptors on the surface of T cells in the peripheral blood and within the resected tumor and lymph node specimens; changes in expression of selected immune markers; survival (overall and recurrence free survival)Recruiting
NCT
03459222
Relatlimab (anti-LAG3 monoclonal antibody BMS-986016) + nivolumab (anti-PD-1) or + BMS-986205 (IDO1 inhibitor) or relatlimab + nivolumab and ipilimumab (anti-CTLA-4)I/IIAdvanced malignant tumorsToxicity, safety, objective response rate, disease control rate, median duration of responseRecruiting
NCT
03867799
Relatlimab (anti-LAG3 monoclonal antibody BMS-986016) + nivolumab (anti-PD-1)IIMetastatic colorectal cancerDisease control rate, toxicity, duration of disease control, best objective response rate, progression free survival, overall survivalNot yet recruiting
NCT
02519322
Nivolumab (anti-PD-1); nivolumab + relatlimab (anti-LAG3 monoclonal antibody BMS-986016); nivolumab + ipilimumab (anti-CTLA-4)IIMelanomaPathologic response; immunological response (changes in T cell infiltrate); objective response; survival (recurrence free and overall survival); adverse eventsRecruiting
NCT
03704077
Relatlimab (anti-LAG3 monoclonal antibody BMS-986016) + nivolumab (anti-PD-1) + paclitaxel; nivolumab + paclitaxel; ramucirumab + paclitaxel; relatlimab + nivolumab; nivolumabIIRecurrent, locally advanced, or metastatic gastric cancer (GC) or gastroesophageal junction (GEJ) adenocarcinomaOverall response rate; toxicity; duration of response; survival (progression free survival and overall survival)Not yet recruiting
NCT
01968109
Relatlimab (anti-LAG3 monoclonal antibody BMS-986016) + nivolumab (anti-PD-1)I/IIAdvanced solid tumorsSafety (adverse events), tolerability and efficacy (objective response rate, disease control rate, duration of response)Recruiting
NCT
03662659
Relatlimab (anti-LAG3 monoclonal antibody BMS-986016) + investigator’s choice chemotherapy; nivolumab (anti-PD-1) + investigator’s choice chemotherapyIIUnresectable, untreated, locally advanced or metastatic gastric or gastroesophageal junction cancerObjective response rate; toxicity; duration of response; survival (overall survival and progression free survival)Recruiting
NCT
03610711
Nivolumab (anti-PD-1) +/- relatlimab (anti-LAG3 monoclonal antibody BMS-986016) + stereotactic radiotherapyIb/IIRecurrent or limited metastatic gastroesophageal cancerChange in the infiltrating CD8+ T cell density; safety; efficacyRecruiting
NCT
02966548
Relatlimab (anti-LAG3 monoclonal antibody BMS-986016) +/- nivolumab (anti-PD-1)IAdvanced solid tumorsSafety, tolerability, and efficacyRecruiting
NCT
02996110
Relatlimab (anti-LAG3 monoclonal antibody BMS-986016) + nivolumab (anti-PD-1); nivolumab + BMS-986205 (IDO-inhibitor); nivolumab + ipilimumab (anti-CTLA-4)IIAdvanced renal cell carcinomaObjective response rate; duration of response; progression free survival rate; safety; tolerabilityRecruiting
NCT
02935634
Relatlimab (anti-LAG3 monoclonal antibody BMS-986016) + nivolumab (anti-PD-1); nivolumab + ipilimumab (anti-CTLA-4); nivolumab and BMS-986205 (IDO-inhibitor)IIAdvanced gastric cancerObjective response rate; duration of response; progression free survival rate; adverse eventsRecruiting
NCT
02488759
Nivolumab (anti-PD-1); nivolumab + ipilimumab (anti-CTLA-4); nivolumab + relatlimab (anti-LAG3 monoclonal antibody BMS-986016); nivolumab + daratumumab (anti-CD38)I/IIVirus associated tumors (anal canal cancer; cervical cancer; Epstein Barr Virus (EBV) positive gastric cancer; HPV positive and negative squamous cell cancer of the head and neck (SCCHN); Merkel cell cancer; nasopharyngeal cancer; penile cancer; vaginal and vulvar cancerSafety, tolerability; objective response rate; survival (progression free survival; overall survival); duration of responseRecruiting
NCT
02750514
Nivolumab (anti-PD-1); nivolumab + dasatinib (src, c-Kit, ephri receptor inhibitor); nivolumab + relatlimab (anti-LAG3 monoclonal antibody BMS-986016); nivolumab + ipilimumab (anti-CTLA-4); nivolumab + BMS-986205 (IDO-inhibitor)IIAdvanced non-small cell lung cancerObjective response; duration of response; progression free survival rate; safety and tolerabilityRecruiting
NCT
03335540
Nivolumab (anti-PD-1) + relatlimab (anti-LAG3 monoclonal antibody BMS-986016); nivolumab + lirilumab (anti-KIR monoclonal antibody); nivolumab + cabiralizumab (anti-CSF1R); nivolumab + ipilimumab (anti-CTLA-4); nivolumab + anti-GITR; nivolumab + BMS-986205 (IDO-inhibitor); nivolumab + radiation therapyISolid tumorsNumber of patients with biopsy specimens; change from baseline in histopatologic and biomarker expression patterns; number of adverse events; Recruiting
NCT
03219268
MGD013 (Anti-PD-1, anti-LAG-3 bispecific DART protein)IUnresectable, locally advanced or metastatic solid tumors of any histologyAdverse events; pharmacokinetics; pharmacodynamics; immunogenicity, and preliminary antitumor activity Recruiting
NCT
03365791
LAG525 (anti-LAG3 IgG4 antibody) + PDR001 (anti-PD-1 IgG4 antibody)IIAdvanced solid and hematological malignanciesClinical benefit rate; progression free survival; overall response rate; time to response; safety and tolerability; duration of response; time to progressionActive, not recruiting
NCT
03499899
LAG525 (anti-LAG3 IgG4 antibody) + PDR001 (anti-PD-1 IgG4 antibody); LAG525 + PDR001 + carboplatin; LAG525 + carboplatinIIAdvanced triple negative breast cancer (first or second line)Overall response rate; duration of response; overall survival; pharmacokinetic parameters; time to response; progression free survival; clinical benefit rate; anti-drug antibodies prevalence at baseline and incidence on treatmentRecruiting
NCT
02460224
LAG525 (anti-LAG3 IgG4 antibody) +/- PDR001 (anti-PD-1 IgG4 antibody)I/IIAdvances malignanciesDose limiting toxicities; overall response rate; area under the curve; concentration of anti-LAG525 and anti-PDR001 antibodies; correlation of PD-L1 and LAG-3 expression; overall response rate; expression of IFN-γ immune-related genes by mRNA profiling; safety; tolerability; progression free survival; duration of response; disease control rateActive, not recruiting
NCT
03742349
LAG525 (anti-LAG3 IgG4 antibody) + PDR001 (anti-PD-1 IgG4 antibody) + NIR178 (adenosine A2A receptor antagonist); LAG525 + PDR001 + capmatinib (c-Met inhibitor); LAG525 + PDR001 + MCS110 (anti-M-CSF antibody); LAG525 + PDR001 + canakinumab (anti-interleukin-1 beta)IbAdult patients with triple negative breast cancerSafety; dose limiting toxicities; best overall response; progression free survival; presence of anti-PDR001. of anti-LAG525, of anti-MCS110, of anti- canakinumab antibodies; pharmacokinetic and pharmacodynamic parametersRecruiting
NCT
03484923
LAG525 (anti-LAG3 IgG4 antibody) + PDR001 (anti-PD-1 IgG4 antibody); PDR001 + capmatinib (c-Met inhibitor); PDR001 + canakinumab (anti-interleukin-1 beta)IIPreviously treated stage III and stage IV melanoma Overall response rate; duration of response; survival (overall survival and progression free survival); disease control rate; prevalence and incidence of anti-drug antibodies; frequency of patients with a favorable biomarker profileRecruiting
NCT
03005782
REGN3767 (anti-LAG3 monoclonal antibody) +/- REGN2810 (anti-PD-1 monoclonal antibody)IAdvanced tumorsSafety; tolerability; activity and pharmacokineticsRecruiting
NCT
03250832
TSR-033 (humanized monoclonal anti-LAG3 IgG4) +/- anti-PD-1 (IgG4 antibody)IAdvanced solid tumorsAdverse events; responses; pharmacokinetic parameters; duration of response; disease control rate; survival (progression free survival; overall survival)Recruiting

Share and Cite

MDPI and ACS Style

Solinas, C.; Migliori, E.; De Silva, P.; Willard-Gallo, K. LAG3: The Biological Processes That Motivate Targeting This Immune Checkpoint Molecule in Human Cancer. Cancers 2019, 11, 1213. https://0-doi-org.brum.beds.ac.uk/10.3390/cancers11081213

AMA Style

Solinas C, Migliori E, De Silva P, Willard-Gallo K. LAG3: The Biological Processes That Motivate Targeting This Immune Checkpoint Molecule in Human Cancer. Cancers. 2019; 11(8):1213. https://0-doi-org.brum.beds.ac.uk/10.3390/cancers11081213

Chicago/Turabian Style

Solinas, Cinzia, Edoardo Migliori, Pushpamali De Silva, and Karen Willard-Gallo. 2019. "LAG3: The Biological Processes That Motivate Targeting This Immune Checkpoint Molecule in Human Cancer" Cancers 11, no. 8: 1213. https://0-doi-org.brum.beds.ac.uk/10.3390/cancers11081213

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop