Skip to main content

Targeting TIM-3 for hematological malignancy: latest updates from the 2022 ASH annual meeting

Abstract

T cell immunoglobulin domain and mucin domain-3 (TIM-3) is an important immune checkpoint (IC) protein in cancer immunosuppression that is considered a novel target for immunotherapy. Moreover, TIM-3, an immuno-myeloid regulator, is highly expressed on the cells of several solid tumors and myeloid leukemia stem cells (LSCs). TIM-3 blockade was shown to have dual effects for directly inhibiting leukemia cells and restoring T cell activation. We summarize several of the latest reports on the role of TIM-3 in immunotherapy for hematological malignancies from the 2022 ASH Annual Meeting (ASH2022).

To the editor,

T cell immunoglobulin domain and mucin domain-3 (TIM-3), is significantly expressed on the T cells of cancer and leukemia patients and has been discovered to be associated with tumor progression [1]. Moreover, malignant cells can also express TIM-3, and it is preferentially expressed on myeloid leukemia stem cells (LSCs) [2]. Thus, TIM-3 is not only considered a T cell exhaustion molecule but also a potential target for myeloid cells as an immuno-myeloid regulator. Anti-TIM-3 antibodies have been shown to have efficacy and be safe for the treatment of advanced solid tumors. Recently, TIM-3 inhibitors have been used to treat acute myeloid leukemia (AML) and myelodysplastic syndromes (MDS) in clinical trials. Here, we summarize several of the latest reports on the role of TIM-3 in immunotherapy for HMs from the 2022 ASH Annual Meeting (ASH2022).

Influence of CAR-T cell activation and BiTE function

Chimeric antigen receptor (CAR) –T cell exhaustion or senescence in vivo remains a major issue. Increased expression of immune checkpoint proteins, such as that of TIM-3 during CART19-28ζ cell exhaustion, results in a decrease in cytotoxicity [3]. Moreover, CART19-BBζ cells are more prone to developing a senescent phenotype compared to CART19-28ζ cells [4].

Bispecific antibodies, such as CD19-CD3 T cell engagers (BiTE), have been used for B cell malignancy immunotherapy [5]. In ASH2022, the multicohort, open-label, phase 1/2 MajesTEC-1 study, which investigated the safety/efficacy of teclistamab (B-cell maturation antigen (BCMA)-CD3 bispecific IgG4 antibody) in patients with relapsed/refractory multiple myeloma (RRMM), demonstrated encouraging efficacy. Lower T cell numbers and a higher frequency of T cells expressing IC markers, including TIM-3, are underlying reasons for non-responders with unfavorable immune characteristics at baseline [6]. Overall, the above studies are consistent with the finding of terminally exhausted PD-1 + Tim-3 + T cells in a mouse model of B-cell tumors [7].

TIM-3 blockade for AML and MDS immunotherapy

The first anti-TIM-3 antibody, sabatolimab (MBG453), demonstrated safety and efficacy in a phase I/Ib clinical trial (NCT02608268) in advanced solid tumors. In the STIMULUS clinical trial (NCT03066648), 53 patients with very high risk/high risk (vHR/HR)-MDS and 48 patients with newly diagnosed (ND) AML were treated with sabatolimab plus hypomethylating agent (HMA). A higher overall response rate (ORR) and 1-year progression-free survival (PFS) were shown for 51 vHR/HR-MDS and 40 ND-AML patients (Table 1). At present, 3 of 11 clinical studies involving TIM-3 inhibitors in AML/MDS have completed recruitment (NCT03066648, NCT03946670, and NCT04266301). In ASH2022, primary results from the ongoing STIMULUS-MDS1 (NCT03946670) were reported [8]. This trial is a randomized, double-blind, placebo-controlled, Ph II study of sabatolimab + HMA in patients with intermediate risk (IR), HR, or vHR-MDS who were ineligible for intensive chemotherapy or hematopoietic stem cell transplantation at screening. A total of 127 patients were randomized to either sabatolimab + HMA or placebo + HMA (Table 1). The primary complete remission (CR) rate was 21.5% vs. 17.7%, respectively, for these two groups. The CR + partial remission (PR) + hematologic improvement (HI) was 49.2% (sabatolimab + HMA) vs. 37.1% (placebo + HMA), respectively. In addition, the ongoing Ph III STIMULUS-MDS2 trial, which has a primary endpoint of overall survival (OS) (NCT04266301), has completed accrual. Another clinical trial of sabatolimab combined with an oral HMA drug (NCT04878432) is underway. Overall, sabatolimab + HMA was associated with a favorable safety profile in patients with HR-MDS [9].

Table 1 The efficacy of sabatolimab in the treatment of AML and MDS

In contrast, TIM-3-CD28 fusion proteins that turn inhibitory signals derived from TIM-3 engagement into activation by CD28 have been designed. These fusion proteins alone demonstrate the strongest response upon stimulation with anti-CD3 antibodies. Importantly, these proteins can increase the proliferation, activation, and cytotoxic capacity of conventional anti-CD19 CAR T cells [10]. Thus, combining IC fusion proteins with anti-CD19 CARs has the potential to increase the T cell proliferation capacity. In 2021, Lee et al. designed second-generation anti-TIM-3 CAR-T cells that exhibit potent anti-AML activity, including primary LSCs. Thus, anti-TIM-3 CAR-T cell therapy might be considered following first-line therapy to eradicate the LSCs present in minimal residual disease.

Targeting the TIM-3 ligand Gal-9 for HM immunotherapy

Targeting the TIM-3 ligand galectin-9 (Gal-9) was recently considered as a novel immunotherapy for HMs. LYT-200, a fully humanized IgG4 αGAL-9 antibody, has been well tolerated in a phase I clinical trial for solid tumors. In ASH2022, a study reported the efficacy of LYT-200 in vitro and in vivo in multiple HMs. Further study demonstrated that LYT-200 also has in vivo protection and survival benefit in murine models of T cell acute lymphoblastic leukemia (T-ALL) and AML [11]. In ASH2022, another investigation with single-cell omics analyses demonstrated that Gal-9 expressed on chronic lymphocytic leukemia (CLL) cells led to T cell exhaustion, and Gal-9 antibody treatment of the CLL mouse model reduced disease development. These findings further supported that Gal-9 can be a novel immunotherapy target for CLL [12].

In summary, targeting TIM-3 has dual effects for directly inhibiting leukemia cells and restoring T cell activation. In addition, Gal-9 may serve as a novel immunotherapeutic target. Several targeted agents against TIM-3 are at different stages of development (Table 2).

Table 2 Targeted agents against TIM-3

Data Availability

The material supporting the conclusion of this study has been included within the article.

Abbreviations

AML:

acute myeloid leukemia

AEs:

adverse events

BiTE:

Bispecific T-cell engagers

BCMA:

B-cell maturation antigen

CLL:

chronic lymphocytic leukemia

CAR:

Chimeric antigen receptor

GAL-9:

galectin-9

HMs:

hematological malignancies

HI:

hematologic improvement

IC:

Immune checkpoint

IR:

intermediate risk

LSCs:

leukemia stem cells

MDS:

myelodysplastic syndromes

ND:

newly diagnosed

ORR:

overall response rate

PFS:

progression-free survival

PR:

partial remission

RRMM:

relapsed/refractory multiple myeloma

T-ALL:

T cell-acute lymphoblastic leukemia

TIM-3:

T cell immunoglobulin domain and mucin domain-3

vHR/HR:

very high risk/high risk

References

  1. Corsale AM, Shekarkar Azgomi M, Plano F, Di Simone M, Perez C, Picone C, Gigliotta E, Vullo C, Camarda G, Rotolo C, Speciale M, Santoro M, Caccamo N, Dieli F, Paiva B, Meraviglia S, Siragusa S, Botta C. High TIM-3 expression may contribute to the functional impairment of bone marrow and circulating Gamma Delta T Lymphocytes during the progression of multiple myeloma. Blood. 2022;140(Supplement 1):9943–4.

    Article  Google Scholar 

  2. Kikushige Y, Shima T, Takayanagi S, Urata S, Miyamoto T, Iwasaki H, Takenaka K, Teshima T, Tanaka T, Inagaki Y, Akashi K. TIM-3 is a promising target to selectively kill acute myeloid leukemia stem cells. Cell Stem Cell. 2010;7(6):708–17.

    Article  CAS  PubMed  Google Scholar 

  3. Stewart CM, Cox MJ, Sakemura RL, Ogbodo EJ, Can I, Manriquez-Roman C, Yun K, Sirpilla O, Girsch JH, Huynh T, Siegler EL, Kim JJ, Mattie M, Scholler N, Filosto S, Kenderian SS. Identification of IL-4 as a Key Regulator of CAR T-Cell exhaustion using Functional Genomics and Correlates of the Zuma-1 clinical trial. Blood. 2022;140(Supplement 1):4536–7.

    Article  Google Scholar 

  4. Can I, Sakemura RL, Manriquez-Roman C, Sirpilla O, Stewart CM, Yun K, Cox MJ, Huynh T, Girsch JH, Ogbodo EJ, Kankeu Fonkoua LA, Hefazi M, Ruff MW, Boysen JC, Siegler EL, Ekiz A, Kenderian SS, Kay NE. Differential susceptibility to Senescence in CART cells based on Co-Stimulatory Signaling. Blood. 2022;140(Supplement 1):8600–1.

    Article  Google Scholar 

  5. Kegyes D, Constantinescu C, Vrancken L, Rasche L, Gregoire C, Tigu B, Gulei D, Dima D, Tanase A, Einsele H, Ciurea S, Tomuleasa C, Caers J. Patient selection for CAR T or BiTE therapy in multiple myeloma: which treatment for each patient? J Hematol Oncol. 2022;15(1):78.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  6. Cortes-Selva D, Casneuf T, Vishwamitra D, Stein S, Perova T, Skerget S, Ramos E, van Steenbergen L, De Maeyer D, Boominathan R, Lau O, Davis C, Banerjee A, Stephenson T, Uhlar CM, Kobos R, Goldberg JD, Pei L, Trancucci D, Girgis S, Wang Lin SX, Wu LS, Moreau P, Usmani S, Bahlis NJ, Van De Donk NW, Verona R. Teclistamab, a B-Cell maturation Antigen (BCMA) x CD3 bispecific antibody, in patients with Relapsed/Refractory multiple myeloma (RRMM): correlative analyses from MajesTEC-1. Blood. 2022;140(Supplement 1):241–3.

    Article  Google Scholar 

  7. Shibamiya A, Mimura N, Miyamoto-Nagai Y, Koide S, Oshima M, Rizq O, Aoyama K, Nakajima-Takagi Y, Kayamori K, Isshiki Y, Oshima-Hasegawa N, Muto T, Tsukamoto S, Takeda Y, Koyama-Nasu R, Chiba T, Honda H, Yokote K, Iwama A, Sakaida E. Exhausted T cells characterized by Upregulation of specific transcription factors are increased in a mouse model of De Novo mature B cell neoplasms. Blood. 2022;140(Supplement 1):4240–1.

    Article  Google Scholar 

  8. Santini V, Platzbecker U, Fenaux P, Giagounidis A, Miyazaki Y, Sekeres MA, Xiao Z, Sanz G, Van Hoef M, Ma F, Hertle S, Marques Ramos P, Zeidan AM. Disease characteristics and International Prognostic Scoring Systems (IPSS, IPSS-R, IPSS-M) in adult patients with higher-risk myelodysplastic syndromes (MDS) participating in two Randomized, Double-Blind, placebo-controlled studies with intravenous Sabatolimab added to Hypomethylating Agents (HMA) (STIMULUS-MDS1 and MDS2). Blood. 2022;140(Supplement 1):1340–2.

    Article  Google Scholar 

  9. Zeidan AM, Ando K, Rauzy O, Turgut M, Wang M-C, Cairoli R, Hou H-A, Kwong Y-L, Arnan Sangerman M, Meers S, Pullarkat VA, Santini V, Malek K, Kiertsman F, Lyu J, Marques Ramos P, Fenaux P, Miyazaki Y, Platzbecker U. Primary results of Stimulus-MDS1: a Randomized, Double-Blind, placebo-controlled phase II study of TIM-3 inhibition with Sabatolimab added to Hypomethylating Agents (HMAs) in adult patients with higher-risk myelodysplastic syndromes (MDS). Blood. 2022;140(Supplement 1):2063–5.

    Article  Google Scholar 

  10. Blaeschke F, Ortner E, Stenger D, Mahdawi J, Apfelbeck A, Habjan N, Weisser T, Kaeuferle T, Willier S, Kobold S, Feuchtinger T. Design and evaluation of TIM-3-CD28 Checkpoint Fusion Proteins to improve Anti-CD19 CAR T-Cell function. Front Immunol. 2022;13:845499.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  11. Henry CJ, Lee M, Filipovic A. Lyt-200, a humanized anti-galectin-9 antibody, exhibits Preclinical Efficacy in Models of Hematological Malignancies. Blood. 2022;140(Supplement 1):8837–7.

    Article  Google Scholar 

  12. Llaó Cid L, Wong JK, Wierz M, Paul Y, Roider T, Fernandez Botana I, Gonder S, Floerchinger A, Colomer D, Campo E, Dietrich S, Lichter P, Zapatka M, Moussay E, Paggetti J, Seiffert M. Single-cell omics analyses identify the TIM-3 ligand Galectin-9 as Novel Immunotherapy Target for chronic lymphocytic leukemia. Blood. 2022;140(Supplement 1):1805–6.

    Article  Google Scholar 

Download references

Acknowledgements

Not applicable.

Funding

The study is supported by grants of the National Natural Science Foundation of China (Nos. 82293630, 82293632, and 82070152).

Author information

Authors and Affiliations

Authors

Contributions

YQL and HT designed the study. YQL and JXT drafted the manuscript. JXT prepared the tables. HT helped to revise the manuscript. All authors participated in the process of drafting and revising the manuscript. All authors read and approved the final manuscript

Corresponding authors

Correspondence to Huo Tan or Yangqiu Li.

Ethics declarations

Competing interests

The authors declare no competing interests.

Ethics approval and consent to participate

This is not applicable for this summary.

Consent for publication

This is not applicable for this summary.

Additional information

Publisher’s Note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Rights and permissions

Open Access This article is licensed under a Creative Commons Attribution 4.0 International License, which permits use, sharing, adaptation, distribution and reproduction in any medium or format, as long as you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons licence, and indicate if changes were made. The images or other third party material in this article are included in the article’s Creative Commons licence, unless indicated otherwise in a credit line to the material. If material is not included in the article’s Creative Commons licence and your intended use is not permitted by statutory regulation or exceeds the permitted use, you will need to obtain permission directly from the copyright holder. To view a copy of this licence, visit http://creativecommons.org/licenses/by/4.0/. The Creative Commons Public Domain Dedication waiver (http://creativecommons.org/publicdomain/zero/1.0/) applies to the data made available in this article, unless otherwise stated in a credit line to the data.

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Tan, J., Tan, H. & Li, Y. Targeting TIM-3 for hematological malignancy: latest updates from the 2022 ASH annual meeting. Exp Hematol Oncol 12, 62 (2023). https://doi.org/10.1186/s40164-023-00421-2

Download citation

  • Received:

  • Accepted:

  • Published:

  • DOI: https://doi.org/10.1186/s40164-023-00421-2

Keywords