1
|
Rajak P. Immune checkpoint inhibitors: From friend to foe. Toxicol Rep 2025; 14:102033. [PMID: 40353246 PMCID: PMC12063143 DOI: 10.1016/j.toxrep.2025.102033] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2024] [Revised: 04/18/2025] [Accepted: 04/20/2025] [Indexed: 05/14/2025] Open
Abstract
Immune checkpoints are crucial in regulating the activation of cell-mediated and humoral immune responses. However, cancer cells hijack this mechanism to evade the immune surveillance and anti-cancer response. Typically, receptors like PD-1 and CTLA4, expressed on immune cells, prevent the activation and differentiation of T cells. They also inhibit the development of autoimmune reactions. However, ligands such as PD-L1 for the receptor PD-1 are also expressed on the surface of cancer cells that help prevent the activation of anti-cancer immune responses by blocking the signalling pathways mediated by PD-1 and CTLA4. Immune checkpoint inhibitors (ICIs) have promising therapeutic efficacy for treating several cancers by activating T cells and their differentiation into effector cells against tumours. Nonetheless, hyperactivated immune cells usually contribute to detrimental issues, also known as immune-related adverse effects (IrAE). IrAEs have been observed in multiple organs, leading to neurological issues, colitis, endocrine dysfunction, renal issues, hepatitis, pneumonitis, and dermatitis. The interplay between hyperactivated T cells and Treg cells helps in orchestrating the development of autoimmunity. Moreover, the crosstalk between proinflammatory interleukins and the development of autoantibodies also mediates the multiorgan effects of ICIs in cancer patients. IrAEs are generally managed by terminating the ICI therapy, reducing the ICI dose, and by using corticosteroids to subvert inflammation. Therefore, the present review aims to delineate the impacts of ICIs on the development of autoimmune diseases and inflammatory outcomes in cancer patients. In addition, mechanistic insight involving immune cells, cytokines, and autoantibodies for ICI-mediated IrAEs will also be discussed with updated findings in this field.
Collapse
Affiliation(s)
- Prem Rajak
- Toxicology Research Laboratory, Department of Animal Science, Kazi Nazrul University, Asansol, West Bengal, India
| |
Collapse
|
2
|
Twomey JD, George S, Zhang B. Fc gamma receptor polymorphisms in antibody therapy: implications for bioassay development to enhance product quality. Antib Ther 2025; 8:87-98. [PMID: 40177643 PMCID: PMC11959696 DOI: 10.1093/abt/tbaf003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2024] [Revised: 01/07/2025] [Accepted: 01/20/2025] [Indexed: 04/05/2025] Open
Abstract
The effectiveness of therapeutic antibodies is often associated with their Fc-mediated effector functions, such as antibody-dependent cellular cytotoxicity and antibody-dependent cellular phagocytosis. These functions rely on interactions between Fc gamma receptors (FcγRs) on immune cells and the Fc region of antibodies. Genetic variations in these receptors, known as FcγR polymorphisms, can influence therapeutic outcomes by altering receptor expression levels, affinity, and function. This review examines the impact of FcγR polymorphisms on antibody therapy, emphasizing their role in developing and optimizing functional bioassays to assess product quality. Understanding these polymorphisms is essential for refining bioassays, which are crucial for accurately characterizing antibody products and ensuring consistency in manufacturing processes.
Collapse
Affiliation(s)
- Julianne D Twomey
- Office of Pharmaceutical Quality Research, Office of Pharmaceutical Quality, Center for Drug Evaluation and Research, Food and Drug Administration, Silver Spring, MD 20993, United States
| | - Sasha George
- Office of Pharmaceutical Quality Research, Office of Pharmaceutical Quality, Center for Drug Evaluation and Research, Food and Drug Administration, Silver Spring, MD 20993, United States
| | - Baolin Zhang
- Office of Pharmaceutical Quality Research, Office of Pharmaceutical Quality, Center for Drug Evaluation and Research, Food and Drug Administration, Silver Spring, MD 20993, United States
| |
Collapse
|
3
|
Musnier A, Corde Y, Verdier A, Cortes M, Pallandre JR, Dumet C, Bouard A, Keskes A, Omahdi Z, Puard V, Poupon A, Bourquard T. AI-enhanced profiling of phage-display-identified anti-TIM3 and anti-TIGIT novel antibodies. Front Immunol 2025; 16:1499810. [PMID: 40134430 PMCID: PMC11933058 DOI: 10.3389/fimmu.2025.1499810] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2024] [Accepted: 02/18/2025] [Indexed: 03/27/2025] Open
Abstract
Antibody discovery is a lengthy and labor-intensive process, requiring extensive laboratory work to ensure that an antibody demonstrates the appropriate efficacy, production, and safety characteristics necessary for its use as a therapeutic agent in human patients. Traditionally, this process begins with phage display or B-cells isolation campaigns, where affinity serves as the primary selection criterion. However, the initial leads identified through this approach lack sufficient characterization in terms of developability and epitope definition, which are typically performed at late stages. In this study, we present a pipeline that integrates early-stage phage display screening with AI-based characterization, enabling more informed decision-making throughout the selection process. Using immune checkpoints TIM3 and TIGIT as targets, we identified five initial leads exhibiting similar binding properties. Two of these leads were predicted to have poor developability profiles due to unfavorable surface physicochemical properties. Of the remaining three candidates, structural models of the complexes formed with their respective targets were generated for 2: T4 (against TIGIT) and 6E9 (against TIM3). The predicted epitopes allowed us to anticipate a competition with TIM3 and TIGIT binding partners, and to infer the antagonistic functions expected from these antibodies. This study lays the foundations of a multidimensional AI-driven selection of lead candidates derived from high throughput analysis.
Collapse
Affiliation(s)
| | | | | | | | - Jean-René Pallandre
- Etablissement Français du Sang - Bourgogne Franche-Comté (EFS BFC), Plateforme ITAC-UMR1098-RIGHT, Besançon, France
| | | | - Adeline Bouard
- Etablissement Français du Sang - Bourgogne Franche-Comté (EFS BFC), Plateforme ITAC-UMR1098-RIGHT, Besançon, France
| | | | | | | | | | | |
Collapse
|
4
|
Zhang M, Yang Y, Liu J, Guo L, Guo Q, Liu W. Bone marrow immune cells and drug resistance in acute myeloid leukemia. Exp Biol Med (Maywood) 2025; 250:10235. [PMID: 40008144 PMCID: PMC11851207 DOI: 10.3389/ebm.2025.10235] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2024] [Accepted: 01/23/2025] [Indexed: 02/27/2025] Open
Abstract
In recent years, the relationship between the immunosuppressive niche of the bone marrow and therapy resistance in acute myeloid leukemia (AML) has become a research focus. The abnormal number and function of immunosuppressive cells, including regulatory T cells (Tregs) and myeloid-derived suppressor cells (MDSCs), along with the dysfunction and exhaustion of immunological effector cells, including cytotoxic T lymphocytes (CTLs), dendritic cells (DCs) and natural killer cells (NKs), can induce immune escape of leukemia cells and are closely linked to therapy resistance in leukemia. This article reviews the research progress on the relationship between immune cells in the marrow microenvironment and chemoresistance in AML, aiming to provide new ideas for the immunotherapy of AML.
Collapse
Affiliation(s)
- Miao Zhang
- Department of Pediatrics (Hematological Oncology), Children Hematological Oncology and Birth Defects Laboratory, The Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan, China
- Sichuan Clinical Research Center for Birth Defects, The Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan, China
| | - You Yang
- Department of Pediatrics (Hematological Oncology), Children Hematological Oncology and Birth Defects Laboratory, The Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan, China
- Sichuan Clinical Research Center for Birth Defects, The Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan, China
| | - Jing Liu
- Department of Pediatrics (Hematological Oncology), Children Hematological Oncology and Birth Defects Laboratory, The Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan, China
- Sichuan Clinical Research Center for Birth Defects, The Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan, China
| | - Ling Guo
- Department of Pediatrics (Hematological Oncology), Children Hematological Oncology and Birth Defects Laboratory, The Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan, China
- Sichuan Clinical Research Center for Birth Defects, The Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan, China
| | - Qulian Guo
- Department of Pediatrics (Hematological Oncology), Children Hematological Oncology and Birth Defects Laboratory, The Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan, China
- Sichuan Clinical Research Center for Birth Defects, The Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan, China
| | - Wenjun Liu
- Department of Pediatrics (Hematological Oncology), Children Hematological Oncology and Birth Defects Laboratory, The Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan, China
- Sichuan Clinical Research Center for Birth Defects, The Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan, China
| |
Collapse
|
5
|
Zhang MG, Gallo RA, Tan CH, Camacho M, Fasih-Ahmad S, Moeyersoms AHM, Sayegh Y, Dubovy SR, Pelaez D, Rong AJ. Single-Cell RNA Profiling of Ocular Adnexal Sebaceous Carcinoma Reveals a Complex Tumor Microenvironment and Identifies New Biomarkers. Am J Ophthalmol 2025; 270:8-18. [PMID: 39393421 PMCID: PMC11735305 DOI: 10.1016/j.ajo.2024.10.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2024] [Revised: 09/23/2024] [Accepted: 10/03/2024] [Indexed: 10/13/2024]
Abstract
PURPOSE Ocular adnexal sebaceous carcinoma (OaSC) is an aggressive malignancy that often necessitates orbital exenteration. Its tumor composition and transcriptional profile remain largely unknown, which poses a significant barrier to medical advances. Here, we report the first in-depth transcriptomic analysis of OaSC at the single-cell resolution and discern mechanisms underlying cancer progression for the discovery of potential globe-sparing immunotherapies, targeted therapies, and biomarkers to guide clinical management. DESIGN Laboratory investigation with a retrospective observational case series. METHODS Single-cell RNA sequencing was performed on six patient specimens: three primary tumors, two tumors with pagetoid spread, and a normal tarsus sample. Cellular components were identified via gene signatures. Molecular pathways underlying tumorigenesis and pagetoid spread were discerned via gene ontology analysis of the differentially expressed genes between specimens. CALML5 immunohistochemistry was performed on an archival cohort of OaSC, squamous cell carcinoma, ocular surface squamous neoplasia (OSSN), and basal cell carcinoma cases. RESULTS Analysis of 29,219 cells from OaSC specimens revealed tumor, immune, and stromal cells. Tumor-infiltrating immune cells include a diversity of cell types, including exhausted T-cell populations. In primary OaSC tumors, mitotic nuclear division and oxidative phosphorylation pathways are upregulated, while lipid biosynthesis and metabolism pathways are downregulated. Epithelial tissue migration pathways are upregulated in tumor cells undergoing pagetoid spread. Single-cell RNA sequencing analyses also revealed that CALML5 is upregulated in OaSC tumor cells. Diffuse nuclear and cytoplasmic CALML5 staining was present in 28 of 28 (100%) OaSC cases. Diffuse nuclear and membranous CALML5 staining was present in 5 of 25 (20%) squamous cell carcinoma and OSSN cases, while diffuse nuclear staining was present in 1 of 12 (8%) basal cell carcinoma cases. CONCLUSIONS This study reveals a complex OaSC tumor microenvironment and confirms that the CALML5 immunohistochemical stain is a sensitive diagnostic marker.
Collapse
Affiliation(s)
- Michelle G Zhang
- From the Dr. Nasser Ibrahim Al-Rashid Orbital Vision Research Center (M.G.Z., R.A.G., A.H.M., D.P., and A.J.R.), Bascom Palmer Eye Institute, University of Miami Miller School of Medicine, Miami, Florida, USA; Sylvester Comprehensive Cancer Center (M.G.Z., R.A.G., D.P., and A.J.R.), University of Miami Miller School of Medicine, Miami, Florida, USA
| | - Ryan A Gallo
- From the Dr. Nasser Ibrahim Al-Rashid Orbital Vision Research Center (M.G.Z., R.A.G., A.H.M., D.P., and A.J.R.), Bascom Palmer Eye Institute, University of Miami Miller School of Medicine, Miami, Florida, USA; Sylvester Comprehensive Cancer Center (M.G.Z., R.A.G., D.P., and A.J.R.), University of Miami Miller School of Medicine, Miami, Florida, USA
| | - Charissa H Tan
- Department of Ophthalmology (C.H.T., M.C., S.F.A., Y.S., and S.R.D.), Florida Lions Ocular Pathology Laboratory, Bascom Palmer Eye Institute, University of Miami Miller School of Medicine, Miami, Florida, USA
| | - Matthew Camacho
- Department of Ophthalmology (C.H.T., M.C., S.F.A., Y.S., and S.R.D.), Florida Lions Ocular Pathology Laboratory, Bascom Palmer Eye Institute, University of Miami Miller School of Medicine, Miami, Florida, USA
| | - Sohaib Fasih-Ahmad
- Department of Ophthalmology (C.H.T., M.C., S.F.A., Y.S., and S.R.D.), Florida Lions Ocular Pathology Laboratory, Bascom Palmer Eye Institute, University of Miami Miller School of Medicine, Miami, Florida, USA
| | - Acadia H M Moeyersoms
- From the Dr. Nasser Ibrahim Al-Rashid Orbital Vision Research Center (M.G.Z., R.A.G., A.H.M., D.P., and A.J.R.), Bascom Palmer Eye Institute, University of Miami Miller School of Medicine, Miami, Florida, USA; Sylvester Comprehensive Cancer Center (M.G.Z., R.A.G., D.P., and A.J.R.), University of Miami Miller School of Medicine, Miami, Florida, USA
| | - Yoseph Sayegh
- Department of Ophthalmology (C.H.T., M.C., S.F.A., Y.S., and S.R.D.), Florida Lions Ocular Pathology Laboratory, Bascom Palmer Eye Institute, University of Miami Miller School of Medicine, Miami, Florida, USA
| | - Sander R Dubovy
- Department of Ophthalmology (C.H.T., M.C., S.F.A., Y.S., and S.R.D.), Florida Lions Ocular Pathology Laboratory, Bascom Palmer Eye Institute, University of Miami Miller School of Medicine, Miami, Florida, USA
| | - Daniel Pelaez
- From the Dr. Nasser Ibrahim Al-Rashid Orbital Vision Research Center (M.G.Z., R.A.G., A.H.M., D.P., and A.J.R.), Bascom Palmer Eye Institute, University of Miami Miller School of Medicine, Miami, Florida, USA; Sylvester Comprehensive Cancer Center (M.G.Z., R.A.G., D.P., and A.J.R.), University of Miami Miller School of Medicine, Miami, Florida, USA
| | - Andrew J Rong
- From the Dr. Nasser Ibrahim Al-Rashid Orbital Vision Research Center (M.G.Z., R.A.G., A.H.M., D.P., and A.J.R.), Bascom Palmer Eye Institute, University of Miami Miller School of Medicine, Miami, Florida, USA; Sylvester Comprehensive Cancer Center (M.G.Z., R.A.G., D.P., and A.J.R.), University of Miami Miller School of Medicine, Miami, Florida, USA; Division of Oculofacial Plastic, Reconstructive, and Orbital Surgery (A.J.R.), Bascom Palmer Eye Institute, University of Miami Miller School of Medicine, Miami, Florida, USA.
| |
Collapse
|
6
|
Mehta AK, Konopleva M. Nontransplant treatment approaches for myeloid neoplasm with mutated TP53. HEMATOLOGY. AMERICAN SOCIETY OF HEMATOLOGY. EDUCATION PROGRAM 2024; 2024:326-334. [PMID: 39644032 DOI: 10.1182/hematology.2024000557] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/09/2024]
Abstract
TP53-mutated myelodysplastic syndrome (MDS) and acute myeloid leukemia (AML) remain a challenging spectrum of clonal myeloid disease with poor prognosis. Recent studies have shown that in AML, MDS, and MDS/AML with biallelic TP53 loss, the TP53-mutated clone becomes dominant. These are highly aggressive diseases that are resistant to most chemotherapies. The latest 2022 International Consensus Classification categorizes these diseases under "myeloid disease with mutated TP53." All treatment approaches have not improved survival rates for this disease. Many newer therapies are on the horizon, including chimeric antigen receptor T/NK-cell therapies, mutated p53 reactivators, Fc fusion protein, and monoclonal antibodies targeting various myeloid antigens. This review summarizes the current approaches for myeloid disease with TP53 mutation and provides an overview of emerging nontransplant approaches.
Collapse
Affiliation(s)
- Ansh K Mehta
- Department of Hematology and Oncology, Jacobi Medical Center/Albert Einstein College of Medicine, Bronx, NY
| | - Marina Konopleva
- Department of Hematology and Oncology, Montefiore Medical Center/Albert Einstein College of Medicine, Bronx, NY
| |
Collapse
|
7
|
Karami R, Fathi M, Jalali P, Hassannia H, Zarei A, Hojjat-Farsangi M, Jadidi F. The emerging role of TIM-3 in colorectal cancer: a promising target for immunotherapy. Expert Opin Ther Targets 2024; 28:1093-1115. [PMID: 39670788 DOI: 10.1080/14728222.2024.2442437] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2024] [Revised: 11/30/2024] [Accepted: 12/10/2024] [Indexed: 12/14/2024]
Abstract
INTRODUCTION Colorectal cancer (CRC) imposes a substantial worldwide health burden, necessitating innovative strategies to enhance therapeutic outcomes. T cell immunoglobulin-3 (Tim-3), an immune checkpoint, enhances immunological tolerance. Tim-3's role in CRC surpasses its conventional function as an indicator of dysfunction in T lymphocytes. AREAS COVERED This review provides an all-inclusive summary of the structural and functional attributes of Tim-3's involvement in the case of CRC. It explores the implications of Tim-3 expression in CRC with regard to tumor progression, clinical characteristics, and therapeutic approaches. Furthermore, it delves into the intricate signaling pathways and molecular mechanisms through which Tim-3 exerts its dual function in both immunity against tumors and immune evasion. EXPERT OPINION Understanding Tim-3's complicated network of interactions in CRC has significant consequences for the development of novel immunotherapeutic strategies targeted toward restoring anti-tumor immune responses and improving patient survival. Tim-3 is an important and valuable target for CRC patient risk classification and treatment because it regulates a complex network of strategies for suppressing immune responses, including causing T cell exhaustion, increasing Treg (regulatory T-cell) proliferation, and altering antigen-presenting cell activity.
Collapse
Affiliation(s)
- Reza Karami
- Immunology Research Centre, Tabriz University of Medical Sciences, Tabriz, Iran
- Department of Immunology, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Mehrdad Fathi
- Immunology Research Centre, Tabriz University of Medical Sciences, Tabriz, Iran
- Department of Immunology, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Pooya Jalali
- Immunology Research Centre, Tabriz University of Medical Sciences, Tabriz, Iran
- Department of Immunology, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Hadi Hassannia
- Department of Paramedicine, Amol School of Paramedical Sciences, Mazandaran University of Medical Sciences, Sari, Iran
| | - Asieh Zarei
- Department of Biochemistry, Faculty of Biological Sciences, Tarbiat Modares University, Tehran, Iran
| | | | - Farhad Jadidi
- Immunology Research Centre, Tabriz University of Medical Sciences, Tabriz, Iran
- Department of Immunology, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
| |
Collapse
|
8
|
Hu Y, Wang Y, Min K, Zhou H, Gao X. The influence of immune checkpoint blockade on the outcomes of allogeneic hematopoietic stem cell transplantation. Front Immunol 2024; 15:1491330. [PMID: 39635535 PMCID: PMC11614800 DOI: 10.3389/fimmu.2024.1491330] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2024] [Accepted: 11/04/2024] [Indexed: 12/07/2024] Open
Abstract
The principle of immune checkpoint blockade therapy is based on the activation of T cells. Immune checkpoint inhibitors (ICIs), such as anti-PD-1/PD-L1 and anti-CTLA-4 antibodies, have demonstrated effectiveness in treating solid tumors by reinvigorating the immune system to recognize and eliminate malignant cells. In recent years, ICIs have shown promise in certain patients with relapsed or refractory lymphoma and myeloid malignancies. Allogeneic hematopoietic stem cell transplant (allo-HCT) currently remains the only curative immunotherapy option for eligible patients with these hematologic malignancies. An increasing number of patients with indications for allo-HCT have received treatment with ICIs either before the procedure or as a therapy for relapse after allo-HCT. Nevertheless, initial reports suggest that patients exposed to immune checkpoint inhibitors either before or after allo-HCT are at an increased risk of developing severe graft-versus-host disease and other immune-related adverse events, likely due to the persistent effects of immune checkpoint blocking. Maximizing therapeutic benefits while minimizing side effects of the combination of checkpoint blockade immunotherapy and allo-HCT is an active area of research aimed at improving the prognosis of relapsed or refractory hematologic malignancies. However, there is still a lack of rational design strategies to optimize the combined use of these two different types of immunotherapies. In this review, we addressed the scientific rationale behind ICIs for treating lymphoma and myeloid malignancies. We also summarized the evidence supporting the use of ICIs as salvage therapy before and after allo-HCT. Additionally, we offered insights into current approaches for preventing and treating graft-versus-host disease and other immune-related adverse events during the procedure.
Collapse
Affiliation(s)
- Yalei Hu
- Senior Department of Hematology, the Fifth Medical Center of PLA General Hospital, Beijing, China
- Graduate School, Chinese PLA General Hospital, Beijing, China
| | - Yuxin Wang
- Senior Department of Hematology, the Fifth Medical Center of PLA General Hospital, Beijing, China
- Graduate School, Chinese PLA General Hospital, Beijing, China
| | - Kaili Min
- Senior Department of Hematology, the Fifth Medical Center of PLA General Hospital, Beijing, China
- Graduate School, Chinese PLA General Hospital, Beijing, China
| | - Huisheng Zhou
- Senior Department of Hematology, the Fifth Medical Center of PLA General Hospital, Beijing, China
- Graduate School, Chinese PLA General Hospital, Beijing, China
| | - Xiaoning Gao
- Senior Department of Hematology, the Fifth Medical Center of PLA General Hospital, Beijing, China
- State Key Laboratory of Experimental Hematology, the Fifth Medical Center of PLA General Hospital, Beijing, China
| |
Collapse
|
9
|
Hochman MJ, DeZern AE. SOHO State of the Art Updates and Next Questions: An Update on Higher Risk Myelodysplastic Syndromes. CLINICAL LYMPHOMA, MYELOMA & LEUKEMIA 2024; 24:573-582. [PMID: 38594129 DOI: 10.1016/j.clml.2024.03.006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/31/2024] [Revised: 03/13/2024] [Accepted: 03/14/2024] [Indexed: 04/11/2024]
Abstract
Higher-risk myelodysplastic syndromes (HR-MDS) are clonal myeloid neoplasms that cause life-limiting complications from severe cytopenias and leukemic transformation. Efforts to better classify, prognosticate, and assess therapeutic responses in HR-MDS have resulted in publication of new clinical tools in the last several years. Given limited current treatment options and suboptimal outcomes, HR-MDS stands to benefit from the study of investigational agents.Higher-risk myelodysplastic syndromes (HR-MDS) are a heterogenous group of clonal myeloid-lineage malignancies often characterized by high-risk genetic lesions, increased blood transfusion needs, constitutional symptoms, elevated risk of progression to acute myeloid leukemia (AML), and therapeutic need for allogeneic bone marrow transplantation. Use of blast percentage and other morphologic features to define myelodysplastic neoplasm subtypes is rapidly shifting to incorporate genetics, resulting in a subset of former HR-MDS patients now being considered as AML in presence of leukemia-defining genetic alterations. A proliferation of prognostic tools has further focused use of genetic features to drive decision making in clinical management. Recently, criteria to assess response of HR-MDS to therapy were revised to incorporate more clinically meaningful endpoints and better match AML response criteria. Basic science investigations have resulted in improved understanding of the relationship between MDS genetic lesions, bone marrow stromal changes, germline predispositions, and disease phenotype. However, therapeutic advances have been more limited. There has been import of the IDH1 inhibitor ivosidenib, initially approved for AML; the Bcl-2 inhibitor venetoclax and liposomal daunorubicin/cytarabine (CPX-351) are under active investigation as well. Unfortunately, effective treatment of TP53-mutated disease remains elusive, though preliminary evidence suggests improved outcomes with oral decitabine/cedazuridine over parenteral hypomethylating agent monotherapy. Investigational agents with novel mechanisms of action may help expand the repertoire of treatment options for HR-MDS and trials continue to offer a hopeful therapeutic avenue for suitable patients.
Collapse
Affiliation(s)
- Michael J Hochman
- Department of Hematology and Medical Oncology, Winship Cancer Institute of Emory University, Atlanta, GA
| | - Amy E DeZern
- Division of Hematological Malignancies, Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University, Baltimore, MD.
| |
Collapse
|
10
|
Grigolo S, Filgueira L. Immunotherapy of Clear-Cell Renal-Cell Carcinoma. Cancers (Basel) 2024; 16:2092. [PMID: 38893211 PMCID: PMC11171115 DOI: 10.3390/cancers16112092] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2024] [Revised: 05/27/2024] [Accepted: 05/29/2024] [Indexed: 06/21/2024] Open
Abstract
Clear-cell Renal-Cell Carcinoma (ccRCC) is the most common type of renal-cell carcinoma (RCC). In many cases, RCC patients manifest the first symptoms during the advanced stage of the disease. For this reason, immunotherapy appears to be one of the dominant treatments to achieve a resolution. In this review, we focus on the presentation of the main immune checkpoint proteins that act as negative regulators of immune responses, such as PD-1, CTLA-4, LAG-3, TIGIT, and TIM-3, and their respective inhibitors. Interleukin-2, another potential component of the treatment of ccRCC patients, has also been covered. The synergy between several immunotherapies is one of the main aspects that unites the conclusions of research in recent years. To date, the combination of several immunotherapies enhances the efficacy of a monotherapy, which often manifests important limitations. Immunotherapy aimed at restoring the anti-cancer immune response in ccRCC, involved in the recognition and elimination of cancer cells, may also be a valid solution for many other types of immunogenic tumors that are diagnosed in the final stages.
Collapse
Affiliation(s)
| | - Luis Filgueira
- Anatomy, University of Fribourg, 1700 Fribourg, Switzerland;
| |
Collapse
|
11
|
Yan Z, Ma T, Wang X, Yi L, Wei P, Zhang H, Wang J. Establishment of novel anti-TIM-3 antibodies interfering with its binding to ligands. Heliyon 2024; 10:e28126. [PMID: 38560237 PMCID: PMC10979056 DOI: 10.1016/j.heliyon.2024.e28126] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2023] [Revised: 03/12/2024] [Accepted: 03/12/2024] [Indexed: 04/04/2024] Open
Abstract
The T cell immunoglobulin and mucin-domain containing-3 (TIM-3) receptor has gained significant attention as a promising target for cancer immunotherapy. The inhibitory effect of T cells by TIM-3 is mediated through the interaction between TIM-3 and its ligands. Ligand-blocking anti-TIM-3 antibodies possess the potential to reactivate antigen-specific T cells and augment anti-tumor immunity. However, the precise ligand-receptor interactions disrupted by the administration of TIM-3 blocking Abs have yet to be fully elucidated. In this study, we have developed a panel of monoclonal antibodies targeting human TIM-3, namely MsT001, MsT065, MsT229, and MsT286. They exhibited high sensitivities (10 pg/mL) and affinities (3.70 × 10-9 to 4.61 × 10-11 M) for TIM-3. The TIM-3 antibodies recognized distinct epitopes, including linear epitopes (MsT001 and MsT065), and a conformational epitope (MsT229 and MsT286). Additionally, the MsT229 and MsT286 displayed reactivity towards cynomolgus TIM-3. The interactions between TIM-3/Gal-9, TIM-3/HMGB-1, and TIM-3/CEACAM-1 disrupt the binding of MsT229 and MsT286, while leaving the binding of MsT001 and MsT065 unaffected. The inhibitory effect on the interaction between Gal-9 and TIM-3 was found to be dose-dependently in the presence of either MsT229 or MsT286. The findings suggested that the involvement of conformational epitopes in TIM-3 is crucial for its interaction with ligands, and we successfully generated novel anti-TIM-3 Abs that exhibit inhibitory potential. In conclusion, our finding offers valuable insights -on the comprehension and targeting of human TIM-3.
Collapse
Affiliation(s)
- Zhuohong Yan
- Cancer Research Center, Beijing Chest Hospital, Capital Medical University, Beijing Tuberculosis and Thoracic Tumor Research Institute, Beijing, 101149, China
| | - Teng Ma
- Cancer Research Center, Beijing Chest Hospital, Capital Medical University, Beijing Tuberculosis and Thoracic Tumor Research Institute, Beijing, 101149, China
| | - Xiaojue Wang
- Cancer Research Center, Beijing Chest Hospital, Capital Medical University, Beijing Tuberculosis and Thoracic Tumor Research Institute, Beijing, 101149, China
| | - Ling Yi
- Cancer Research Center, Beijing Chest Hospital, Capital Medical University, Beijing Tuberculosis and Thoracic Tumor Research Institute, Beijing, 101149, China
| | - Panjian Wei
- Cancer Research Center, Beijing Chest Hospital, Capital Medical University, Beijing Tuberculosis and Thoracic Tumor Research Institute, Beijing, 101149, China
| | - Hongtao Zhang
- Cancer Research Center, Beijing Chest Hospital, Capital Medical University, Beijing Tuberculosis and Thoracic Tumor Research Institute, Beijing, 101149, China
| | - Jinghui Wang
- Cancer Research Center, Beijing Chest Hospital, Capital Medical University, Beijing Tuberculosis and Thoracic Tumor Research Institute, Beijing, 101149, China
- Department of Medical Oncology, Beijing Chest Hospital, Capital Medical University, Beijing Tuberculosis and Thoracic Tumor Research Institute, Beijing, 101149, China
| |
Collapse
|
12
|
Brunner AM, Esteve J, Porkka K, Knapper S, Traer E, Scholl S, Garcia-Manero G, Vey N, Wermke M, Janssen JJWM, Narayan R, Fleming S, Loo S, Tovar N, Kontro M, Ottmann OG, Naidu P, Sun H, Han M, White R, Zhang N, Mohammed A, Sabatos-Peyton CA, Steensma DP, Rinne ML, Borate UM, Wei AH. Phase Ib study of sabatolimab (MBG453), a novel immunotherapy targeting TIM-3 antibody, in combination with decitabine or azacitidine in high- or very high-risk myelodysplastic syndromes. Am J Hematol 2024; 99:E32-E36. [PMID: 37994196 DOI: 10.1002/ajh.27161] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2023] [Revised: 10/13/2023] [Accepted: 11/02/2023] [Indexed: 11/24/2023]
Abstract
The safety and efficacy of sabatolimab, a novel immunotherapy targeting T-cell immunoglobulin domain and mucin domain-3 (TIM-3), was assessed in combination with hypomethylating agents (HMAs) in patients with HMA-naive revised International Prognostic System Score (IPSS-R) high- or very high-risk myelodysplastic syndromes (HR/vHR-MDS) or chronic myelomonocytic leukemia (CMML). Sabatolimab + HMA had a safety profile similar to that reported for HMA alone and demonstrated durable clinical responses in patients with HR/vHR-MDS. These results support the ongoing evaluation of sabatolimab-based combination therapy in MDS, CMML, and acute myeloid leukemia.
Collapse
Affiliation(s)
| | | | - Kimmo Porkka
- Helsinki University Hospital Comprehensive Cancer Center, Helsinki, Finland
| | | | - Elie Traer
- Oregon Health & Science University, Portland, Oregon, USA
| | | | | | | | - Martin Wermke
- TU Dresden, NCT/UCC Early Clinical Trial Unit, Dresden, Germany
| | | | - Rupa Narayan
- Massachusetts General Hospital, Boston, Massachusetts, USA
| | | | - Sun Loo
- The Alfred Hospital, Melbourne, Victoria, Australia
| | | | - Mika Kontro
- Helsinki University Hospital Comprehensive Cancer Center, Helsinki, Finland
| | | | | | - Haiying Sun
- Novartis Pharmaceuticals Corporation, East Hanover, New Jersey, USA
| | - May Han
- Cure Ventures, Boston, Massachusetts, USA
| | | | - Na Zhang
- Novartis Institutes for BioMedical Research, Cambridge, Massachusetts, USA
| | - Anisa Mohammed
- Novartis Pharmaceuticals Corporation, East Hanover, New Jersey, USA
| | | | - David P Steensma
- Novartis Institutes for BioMedical Research, Cambridge, Massachusetts, USA
| | | | - Uma M Borate
- Oregon Health & Science University, Portland, Oregon, USA
| | - Andrew H Wei
- The Peter MacCallum Cancer Centre and Royal Melbourne Hospital Walter and Eliza Hall Institute of Medical Research, Melbourne, Victoria, Australia
| |
Collapse
|
13
|
Merz AMA, Platzbecker U. Beyond the horizon: emerging therapeutic approaches in myelodysplastic neoplasms. Exp Hematol 2024; 130:104130. [PMID: 38036096 DOI: 10.1016/j.exphem.2023.11.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2023] [Revised: 10/28/2023] [Accepted: 11/06/2023] [Indexed: 12/02/2023]
Abstract
Management of myelodysplastic neoplasms (MDS) requires a personalized approach, with a focus on improving quality of life and extending lifespan. The International Prognostic Scoring System-Revised and the molecular International Prognostic Scoring System are key tools for risk stratification and management of MDS. They provide a framework for predicting survival and the risk of transformation to acute myeloid leukemia. However, a major challenge in MDS management remains the limited therapeutic options available, especially after the failure of first-line therapies. In lower-risk MDS, the failure of erythropoietin-stimulating agents often leaves few alternatives, although in higher-risk MDS, the prognosis after hypomethylating agent failure is dismal. This highlights the urgent need for novel, more personalized therapeutic approaches. In this review, we discuss emerging novel therapeutic approaches in the treatment of MDS. Several new therapeutic targets are currently being evaluated, offering hope for improved management of MDS in the future.
Collapse
Affiliation(s)
- Almuth Maria Anni Merz
- Department of Hematology, Cellular Therapy, Hemostaseology and Infectious Disease, University Hospital of Leipzig, University of Leipzig Faculty of Medicine Leipzig, Germany.
| | - Uwe Platzbecker
- Department of Hematology, Cellular Therapy, Hemostaseology and Infectious Disease, University Hospital of Leipzig, University of Leipzig Faculty of Medicine Leipzig, Germany.
| |
Collapse
|
14
|
Meng L, Wu H, Wu J, Ding P, He J, Sang M, Liu L. Mechanisms of immune checkpoint inhibitors: insights into the regulation of circular RNAS involved in cancer hallmarks. Cell Death Dis 2024; 15:3. [PMID: 38177102 PMCID: PMC10766988 DOI: 10.1038/s41419-023-06389-5] [Citation(s) in RCA: 50] [Impact Index Per Article: 50.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2023] [Revised: 12/06/2023] [Accepted: 12/11/2023] [Indexed: 01/06/2024]
Abstract
Current treatment strategies for cancer, especially advanced cancer, are limited and unsatisfactory. One of the most substantial advances in cancer therapy, in the last decades, was the discovery of a new layer of immunotherapy approach, immune checkpoint inhibitors (ICIs), which can specifically activate immune cells by targeting immune checkpoints. Immune checkpoints are a type of immunosuppressive molecules expressed on immune cells, which can regulate the degree of immune activation and avoid autoimmune responses. ICIs, such as anti-PD-1/PD-L1 drugs, has shown inspiring efficacy and broad applicability across various cancers. Unfortunately, not all cancer patients benefit remarkably from ICIs, and the overall response rates to ICIs remain relatively low for most cancer types. Moreover, the primary and acquired resistance to ICIs pose serious challenges to the clinical application of cancer immunotherapy. Thus, a deeper understanding of the molecular biological properties and regulatory mechanisms of immune checkpoints is urgently needed to improve clinical options for current therapies. Recently, circular RNAs (circRNAs) have attracted increasing attention, not only due to their involvement in various aspects of cancer hallmarks, but also for their impact on immune checkpoints in shaping the tumor immune microenvironment. In this review, we systematically summarize the current status of immune checkpoints in cancer and the existing regulatory roles of circRNAs on immune checkpoints. Meanwhile, we also aim to settle the issue in an evidence-oriented manner that circRNAs involved in cancer hallmarks regulate the effects and resistance of ICIs by targeting immune checkpoints.
Collapse
Affiliation(s)
- Lingjiao Meng
- Department of Tumor Immunotherapy, The Fourth Hospital of Hebei Medical University, Shijiazhuang, Hebei, 050035, China
- Research Center and Tumor Research Institute, The Fourth Hospital of Hebei Medical University, Shijiazhuang, Hebei, 050017, China
| | - Haotian Wu
- The Third Department of Surgery, The Fourth Hospital of Hebei Medical University, Shijiazhuang, Hebei, 050011, China
- Hebei Key Laboratory of Precision Diagnosis and Comprehensive Treatment of Gastric Cancer, Shijiazhuang, 050011, China
| | - Jiaxiang Wu
- The Third Department of Surgery, The Fourth Hospital of Hebei Medical University, Shijiazhuang, Hebei, 050011, China
- Hebei Key Laboratory of Precision Diagnosis and Comprehensive Treatment of Gastric Cancer, Shijiazhuang, 050011, China
| | - Ping'an Ding
- The Third Department of Surgery, The Fourth Hospital of Hebei Medical University, Shijiazhuang, Hebei, 050011, China
- Hebei Key Laboratory of Precision Diagnosis and Comprehensive Treatment of Gastric Cancer, Shijiazhuang, 050011, China
| | - Jinchen He
- The Third Department of Surgery, The Fourth Hospital of Hebei Medical University, Shijiazhuang, Hebei, 050011, China
- Hebei Key Laboratory of Precision Diagnosis and Comprehensive Treatment of Gastric Cancer, Shijiazhuang, 050011, China
| | - Meixiang Sang
- Research Center and Tumor Research Institute, The Fourth Hospital of Hebei Medical University, Shijiazhuang, Hebei, 050017, China.
- Science and Education Department, Shanghai Electric Power Hospital, Shanghai, 20050, China.
| | - Lihua Liu
- Department of Tumor Immunotherapy, The Fourth Hospital of Hebei Medical University, Shijiazhuang, Hebei, 050035, China.
| |
Collapse
|
15
|
Huuhtanen J, Adnan-Awad S, Theodoropoulos J, Forstén S, Warfvinge R, Dufva O, Bouhlal J, Dhapola P, Duàn H, Laajala E, Kasanen T, Klievink J, Ilander M, Jaatinen T, Olsson-Strömberg U, Hjorth-Hansen H, Burchert A, Karlsson G, Kreutzman A, Lähdesmäki H, Mustjoki S. Single-cell analysis of immune recognition in chronic myeloid leukemia patients following tyrosine kinase inhibitor discontinuation. Leukemia 2024; 38:109-125. [PMID: 37919606 PMCID: PMC10776410 DOI: 10.1038/s41375-023-02074-w] [Citation(s) in RCA: 9] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2023] [Revised: 09/19/2023] [Accepted: 10/17/2023] [Indexed: 11/04/2023]
Abstract
Immunological control of residual leukemia cells is thought to occur in patients with chronic myeloid leukemia (CML) that maintain treatment-free remission (TFR) following tyrosine kinase inhibitor (TKI) discontinuation. To study this, we analyzed 55 single-cell RNA and T cell receptor (TCR) sequenced samples (scRNA+TCRαβ-seq) from patients with CML (n = 13, N = 25), other cancers (n = 28), and healthy (n = 7). The high number and active phenotype of natural killer (NK) cells in CML separated them from healthy and other cancers. Most NK cells in CML belonged to the active CD56dim cluster with high expression of GZMA/B, PRF1, CCL3/4, and IFNG, with interactions with leukemic cells via inhibitory LGALS9-TIM3 and PVR-TIGIT interactions. Accordingly, upregulation of LGALS9 was observed in CML target cells and TIM3 in NK cells when co-cultured together. Additionally, we created a classifier to identify TCRs targeting leukemia-associated antigen PR1 and quantified anti-PR1 T cells in 90 CML and 786 healthy TCRβ-sequenced samples. Anti-PR1 T cells were more prevalent in CML, enriched in bone marrow samples, and enriched in the mature, cytotoxic CD8 + TEMRA cluster, especially in a patient maintaining TFR. Our results highlight the role of NK cells and anti-PR1 T cells in anti-leukemic immune responses in CML.
Collapse
Affiliation(s)
- Jani Huuhtanen
- Translational Immunology Research Program and Department of Clinical Chemistry and Hematology, University of Helsinki, Helsinki, Finland.
- Hematology Research Unit Helsinki, Helsinki University Hospital Comprehensive Cancer Center, Helsinki, Finland.
- Department of Computer Science, Aalto University, Espoo, Finland.
- iCAN Digital Precision Cancer Medicine Flagship, Helsinki, Finland.
| | - Shady Adnan-Awad
- Translational Immunology Research Program and Department of Clinical Chemistry and Hematology, University of Helsinki, Helsinki, Finland
- Hematology Research Unit Helsinki, Helsinki University Hospital Comprehensive Cancer Center, Helsinki, Finland
- iCAN Digital Precision Cancer Medicine Flagship, Helsinki, Finland
- Foundation for the Finnish Cancer Institute, Helsinki, Finland
| | - Jason Theodoropoulos
- Translational Immunology Research Program and Department of Clinical Chemistry and Hematology, University of Helsinki, Helsinki, Finland
- Hematology Research Unit Helsinki, Helsinki University Hospital Comprehensive Cancer Center, Helsinki, Finland
- iCAN Digital Precision Cancer Medicine Flagship, Helsinki, Finland
| | - Sofia Forstén
- Translational Immunology Research Program and Department of Clinical Chemistry and Hematology, University of Helsinki, Helsinki, Finland
- Hematology Research Unit Helsinki, Helsinki University Hospital Comprehensive Cancer Center, Helsinki, Finland
- iCAN Digital Precision Cancer Medicine Flagship, Helsinki, Finland
| | - Rebecca Warfvinge
- Division of Molecular Hematology, Lund Stem Cell Center, Lund University, Lund, Sweden
| | - Olli Dufva
- Translational Immunology Research Program and Department of Clinical Chemistry and Hematology, University of Helsinki, Helsinki, Finland
- Hematology Research Unit Helsinki, Helsinki University Hospital Comprehensive Cancer Center, Helsinki, Finland
- iCAN Digital Precision Cancer Medicine Flagship, Helsinki, Finland
| | - Jonas Bouhlal
- Translational Immunology Research Program and Department of Clinical Chemistry and Hematology, University of Helsinki, Helsinki, Finland
- Hematology Research Unit Helsinki, Helsinki University Hospital Comprehensive Cancer Center, Helsinki, Finland
- iCAN Digital Precision Cancer Medicine Flagship, Helsinki, Finland
| | - Parashar Dhapola
- Division of Molecular Hematology, Lund Stem Cell Center, Lund University, Lund, Sweden
| | - Hanna Duàn
- Translational Immunology Research Program and Department of Clinical Chemistry and Hematology, University of Helsinki, Helsinki, Finland
- Hematology Research Unit Helsinki, Helsinki University Hospital Comprehensive Cancer Center, Helsinki, Finland
- iCAN Digital Precision Cancer Medicine Flagship, Helsinki, Finland
| | - Essi Laajala
- Translational Immunology Research Program and Department of Clinical Chemistry and Hematology, University of Helsinki, Helsinki, Finland
- Hematology Research Unit Helsinki, Helsinki University Hospital Comprehensive Cancer Center, Helsinki, Finland
- iCAN Digital Precision Cancer Medicine Flagship, Helsinki, Finland
| | - Tiina Kasanen
- Translational Immunology Research Program and Department of Clinical Chemistry and Hematology, University of Helsinki, Helsinki, Finland
- Hematology Research Unit Helsinki, Helsinki University Hospital Comprehensive Cancer Center, Helsinki, Finland
- iCAN Digital Precision Cancer Medicine Flagship, Helsinki, Finland
| | - Jay Klievink
- Translational Immunology Research Program and Department of Clinical Chemistry and Hematology, University of Helsinki, Helsinki, Finland
- Hematology Research Unit Helsinki, Helsinki University Hospital Comprehensive Cancer Center, Helsinki, Finland
- iCAN Digital Precision Cancer Medicine Flagship, Helsinki, Finland
| | - Mette Ilander
- Translational Immunology Research Program and Department of Clinical Chemistry and Hematology, University of Helsinki, Helsinki, Finland
- Hematology Research Unit Helsinki, Helsinki University Hospital Comprehensive Cancer Center, Helsinki, Finland
| | - Taina Jaatinen
- Histocompatibility Testing Laboratory, Finnish Red Cross Blood Service, Helsinki, Finland
| | - Ulla Olsson-Strömberg
- Department of Medical Sciences, Uppsala University and Hematology Section, Uppsala University Hospital, Uppsala, Sweden
| | - Henrik Hjorth-Hansen
- Department of Hematology, St. Olavs Hospital, Trondheim, Norway
- Department of Cancer Research and Molecular Medicine, Norwegian University of Science and Technology (NTNU), Trondheim, Norway
| | - Andreas Burchert
- Department of Hematology, Oncology and Immunology, Philipps University Marburg, and University Medical Center Giessen and Marburg, Marburg, Germany
| | - Göran Karlsson
- Division of Molecular Hematology, Lund Stem Cell Center, Lund University, Lund, Sweden
| | - Anna Kreutzman
- Translational Immunology Research Program and Department of Clinical Chemistry and Hematology, University of Helsinki, Helsinki, Finland
- Hematology Research Unit Helsinki, Helsinki University Hospital Comprehensive Cancer Center, Helsinki, Finland
| | - Harri Lähdesmäki
- Department of Computer Science, Aalto University, Espoo, Finland
| | - Satu Mustjoki
- Translational Immunology Research Program and Department of Clinical Chemistry and Hematology, University of Helsinki, Helsinki, Finland.
- Hematology Research Unit Helsinki, Helsinki University Hospital Comprehensive Cancer Center, Helsinki, Finland.
- iCAN Digital Precision Cancer Medicine Flagship, Helsinki, Finland.
| |
Collapse
|
16
|
Zeidan AM, Ando K, Rauzy O, Turgut M, Wang MC, Cairoli R, Hou HA, Kwong YL, Arnan M, Meers S, Pullarkat V, Santini V, Malek K, Kiertsman F, Niolat J, Ramos PM, Menssen HD, Fenaux P, Miyazaki Y, Platzbecker U. Sabatolimab plus hypomethylating agents in previously untreated patients with higher-risk myelodysplastic syndromes (STIMULUS-MDS1): a randomised, double-blind, placebo-controlled, phase 2 trial. Lancet Haematol 2024; 11:e38-e50. [PMID: 38065203 DOI: 10.1016/s2352-3026(23)00333-2] [Citation(s) in RCA: 26] [Impact Index Per Article: 26.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2023] [Revised: 10/18/2023] [Accepted: 10/19/2023] [Indexed: 12/24/2023]
Abstract
BACKGROUND Sabatolimab is an immunotherapy targeting T-cell immunoglobulin domain and mucin domain-3 (TIM-3), an immuno-myeloid regulator expressed on immune cells and leukaemic stem cells. In this trial, we compared the efficacy and safety of sabatolimab plus hypomethylating agent with placebo plus hypomethylating agents in previously untreated patients with higher-risk myelodysplastic syndromes. METHODS STIMULUS-MDS1 was a multicentre, randomised, double-blind, placebo-controlled, phase 2 study done at 54 investigational sites in 17 countries. Adult patients (aged ≥18 years) with intermediate-risk, high-risk, and very high-risk myelodysplastic syndromes (according to Revised International Prognostic Scoring System criteria) who had not received previous treatment were included. Patients were randomly assigned (1:1) to intravenous sabatolimab (400 mg on day 8 and 22) or placebo plus a hypomethylating agent (intravenous decitabine 20 mg/m2 on day 1-5 or intravenous or subcutaneous azacitidine 75 mg/m2 on day 1-7 or day 1-5 and day 8 and 9) every 28 days until treatment discontinuation. The two primary endpoints were complete response rate and progression-free survival, assessed in the full analysis set, which included all randomly assigned patients. Complete response was analysed, as prespecified, 7 months after the last patient was randomly assigned. All other analyses presented, including progression-free survival, were done at the final data cutoff prespecified via a protocol amendment on Sept 2, 2021. Safety was assessed in in all patients who received at least one dose of study treatment. This study is registered with ClinicalTrials.gov, NCT03946670, and is ongoing. FINDINGS Between July 29, 2019, and Aug 10, 2020, 127 patients were randomly assigned to sabatolimab plus a hypomethylating agent group (sabatolimab group; n=65) or placebo plus a hypomethylating agent (placebo group; n=62). The median age of participants was 73 years (IQR 69-77), of whom 86 (68%) of 127 patients were male and 77 (61%) were White. The primary endpoints were not met. Complete response (cutoff date of March 10, 2021) was achieved in 14 (22%; 95% CI 12·3-33·5) of 65 patients in the sabatolimab group vs 11 (18%; 9·2-29·5) of 62 patients in the placebo group (p=0·77). At the cutoff date of the final analysis (March 1, 2022), median follow-up for progression-free survival was 17·8 months (IQR 16·6-19·4) in the sabatolimab group and 19·2 months (17·7-22·3) in the placebo group, and the median progression-free survival was 11·1 months (95% CI 7·6-17·6) in the sabatolimab group vs 8·5 months (6·9-11·3) in the placebo group (hazard ratio 0·75 [95% CI 0·48-1·17]; p=0·1022). The most common adverse events of any grade were neutropenia (35 [56%] of 62 patients in the sabatolimab group vs 43 [68%] of 63 patients in the placebo group), thrombocytopenia (30 [48%] vs 32 [51%]), constipation (29 [47%] vs 24 [38%]), diarrhoea (27 [44%] vs 14 [22%]), anaemia (22 [35%] vs 34 [54%]), febrile neutropenia (22 [35%] vs 15 [24%]), and leukopenia (15 [24%] vs 20 [32%]). One patient developed a serious potential treatment-related immune-mediated adverse event in the sabatolimab group. There was one treatment-related death in the sabatolimab group due to pneumonitis. INTERPRETATION The addition of sabatolimab to hypomethylating agents in this study did not result in a significant improvement in complete response rates or progression-free survival. Sabatolimab had a manageable safety in most patients with higher-risk myelodysplastic syndromes. A randomised phase 3 trial is ongoing to assess the potential benefit of sabatolimab plus azacitidine on overall survival in this setting. FUNDING Novartis Pharmaceuticals.
Collapse
Affiliation(s)
- Amer M Zeidan
- Yale Cancer Center, Yale University, New Haven, CT, USA.
| | - Kiyoshi Ando
- Tokai University School of Medicine, Isehara, Japan
| | - Odile Rauzy
- Institut Universitaire du Cancer Toulouse Oncopole, Toulouse University Hospital, Toulouse, France
| | | | - Ming-Chung Wang
- Kaohsiung-Chang Gung Memorial Hospital, Kaohsiung City, Taiwan
| | - Roberto Cairoli
- ASST Niguarda Hospital, Milan, Italy; Università degli Studi di Milano Bicocca, Milan, Italy
| | - Hsin-An Hou
- National Taiwan University Hospital, Taipei, Taiwan
| | - Yok-Lam Kwong
- University of Hong Kong, Hong Kong Special Administrative Region, China
| | - Montserrat Arnan
- Institut Català d'Oncologia-Hospital Duran i Reynals, IDIBELL, Hospitalet Llobregat, Barcelona, Spain
| | | | | | | | | | | | | | | | | | - Pierre Fenaux
- Hôpital Saint-Louis, Université Paris Cité, Paris, France
| | | | | |
Collapse
|
17
|
Patel SA, Cerny J, Gerber WK, Ramanathan M, Ediriwickrema A, Tanenbaum B, Hutchinson L, Meng X, Flahive J, Barton B, Gillis‐Smith AJ, Suzuki S, Khedr S, Selove W, Higgins AW, Miron PM, Simin K, Woda B, Gerber JM. Prognostic heterogeneity and clonal dynamics within distinct subgroups of myelodysplastic syndrome and acute myeloid leukemia with TP53 disruptions. EJHAEM 2023; 4:1059-1070. [PMID: 38024632 PMCID: PMC10660125 DOI: 10.1002/jha2.791] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 08/10/2023] [Revised: 09/01/2023] [Accepted: 09/04/2023] [Indexed: 12/01/2023]
Abstract
TP53 aberrations constitute the highest risk subset of myelodysplastic neoplasms (MDS) and acute myeloid leukemia (AML). The International Consensus Classification questions the blast threshold between MDS and AML. In this study, we assess the distinction between MDS and AML for 76 patients with TP53 aberrations. We observed no significant differences between MDS and AML regarding TP53 genomics. Median overall survival (OS) was 223 days for the entire group, but prognostic discrimination within subgroups showed the most inferior OS (46 days) for AML with multihit allelic state plus TP53 variant allele frequency (VAF) > 50%. In multivariate analysis, unadjusted Cox models revealed the following variables as independent risk factors for mortality: AML (vs. MDS) (hazard ratio [HR]: 2.50, confidence interval [CI]: 1.4-4.4, p = 0.001), complex karyotype (HR: 3.00, CI: 1.4-6.1, p = 0.003), multihit status (HR: 2.30, CI 1.3-4.2, p = 0.005), and absence of hematopoietic cell transplant (HCT) (HR: 3.90, CI: 1.8-8.9, p = 0.0009). Clonal dynamic modeling showed a significant reduction in TP53 VAF with front-line hypomethylating agents. These findings clarify the impact of specific covariates on outcomes of TP53-aberrant myeloid neoplasms, irrespective of the diagnosis of MDS versus AML, and may influence HCT decisions.
Collapse
Affiliation(s)
- Shyam A. Patel
- Division of Hematology and Oncology, Department of MedicineUMass Memorial Medical Center, UMass Chan Medical SchoolWorcesterMassachusettsUnited States
| | - Jan Cerny
- Division of Hematology and Oncology, Department of MedicineUMass Memorial Medical Center, UMass Chan Medical SchoolWorcesterMassachusettsUnited States
| | - William K. Gerber
- Division of Hematology and Oncology, Department of MedicineUMass Memorial Medical Center, UMass Chan Medical SchoolWorcesterMassachusettsUnited States
| | - Muthalagu Ramanathan
- Division of Hematology and Oncology, Department of MedicineUMass Memorial Medical Center, UMass Chan Medical SchoolWorcesterMassachusettsUnited States
| | - Asiri Ediriwickrema
- Institute for Stem Cell Biology & Regenerative Medicine; Division of Hematology, Department of MedicineStanford UniversityStanfordCaliforniaUnited States
| | - Benjamin Tanenbaum
- Division of Hematology and Oncology, Department of MedicineUMass Memorial Medical Center, UMass Chan Medical SchoolWorcesterMassachusettsUnited States
| | - Lloyd Hutchinson
- Department of PathologyUMass Memorial Medical Center, UMass Chan Medical SchoolWorcesterMassachusettsUnited States
| | - Xiuling Meng
- Department of PathologyUMass Memorial Medical Center, UMass Chan Medical SchoolWorcesterMassachusettsUnited States
| | - Julie Flahive
- Department of Population & Quantitative Health SciencesUMass Chan Medical SchoolWorcesterMassachusettsUnited States
| | - Bruce Barton
- Department of Population & Quantitative Health SciencesUMass Chan Medical SchoolWorcesterMassachusettsUnited States
| | - Andrew J. Gillis‐Smith
- Division of Hematology and Oncology, Department of MedicineUMass Memorial Medical Center, UMass Chan Medical SchoolWorcesterMassachusettsUnited States
| | - Sakiko Suzuki
- Division of Hematology and Oncology, Department of MedicineUMass Memorial Medical Center, UMass Chan Medical SchoolWorcesterMassachusettsUnited States
| | - Salwa Khedr
- Department of PathologyUMass Memorial Medical Center, UMass Chan Medical SchoolWorcesterMassachusettsUnited States
| | - William Selove
- Department of PathologyUMass Memorial Medical Center, UMass Chan Medical SchoolWorcesterMassachusettsUnited States
| | - Anne W. Higgins
- Department of PathologyUMass Memorial Medical Center, UMass Chan Medical SchoolWorcesterMassachusettsUnited States
| | - Patricia M. Miron
- Department of PathologyUMass Memorial Medical Center, UMass Chan Medical SchoolWorcesterMassachusettsUnited States
| | - Karl Simin
- Dept. of MolecularCell & Cancer BiologyUMass Chan Medical SchoolWorcesterMassachusettsUnited States
| | - Bruce Woda
- Department of PathologyUMass Memorial Medical Center, UMass Chan Medical SchoolWorcesterMassachusettsUnited States
| | - Jonathan M. Gerber
- Division of Hematology and Oncology, Department of MedicineUMass Memorial Medical Center, UMass Chan Medical SchoolWorcesterMassachusettsUnited States
| |
Collapse
|
18
|
Tsumura A, Levis D, Tuscano JM. Checkpoint inhibition in hematologic malignancies. Front Oncol 2023; 13:1288172. [PMID: 37920162 PMCID: PMC10619902 DOI: 10.3389/fonc.2023.1288172] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2023] [Accepted: 10/04/2023] [Indexed: 11/04/2023] Open
Abstract
Checkpoint inhibitor therapy has emerged as an effective therapeutic strategy for many types of malignancies, especially in solid tumors. Within the last two decades, numerous monoclonal antibody drugs targeting the CTLA-4 and PD-1/PD-L1 checkpoint pathways have seen FDA approval. Within hematologic malignancies, Hodgkin Lymphoma has seen the greatest clinical benefits thus far with more recent data showing efficacy in the front-line setting. As our understanding of checkpoint inhibition expands, using these pathways as a therapeutic target has shown some utility in the treatment of other hematologic malignancies as well, primarily in the relapsed/refractory settings. Checkpoint inhibition also appears to have a role as a synergistic agent to augment clinical responses to other forms of therapy such as hematopoietic stem cell transplant. Moreover, alternative checkpoint molecules that bypass the well-studied CTLA-4 and PD-1/PD-L1 pathways have emerged as exciting new therapeutic targets. Most excitingly is the use of anti-CD47 blockade in the treatment of high risk MDS and TP-53 mutated AML. Overall, there has been tremendous progress in understanding the benefits of checkpoint inhibition in hematologic malignancies, but further studies are needed in all areas to best utilize these agents. This is a review of the most recent developments and progress in Immune Checkpoint Inhibition in Hematologic Malignancies in the last decade.
Collapse
Affiliation(s)
- Aaron Tsumura
- Division of Malignant Hematology/Cellular Therapy and Transplantation, University of California Davis, Sacramento, CA, United States
| | - Daniel Levis
- School of Medicine, University of California Davis, Sacramento, CA, United States
| | - Joseph M. Tuscano
- Division of Malignant Hematology/Cellular Therapy and Transplantation, University of California Davis, Sacramento, CA, United States
- School of Medicine, University of California Davis, Sacramento, CA, United States
| |
Collapse
|
19
|
Testa U, Castelli G, Pelosi E. TP53-Mutated Myelodysplasia and Acute Myeloid Leukemia. Mediterr J Hematol Infect Dis 2023; 15:e2023038. [PMID: 37435040 PMCID: PMC10332352 DOI: 10.4084/mjhid.2023.038] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2023] [Accepted: 06/01/2023] [Indexed: 07/13/2023] Open
Abstract
TP53-mutated myelodysplastic syndrome (MDS) and acute myeloid leukemia (AML) form a distinct and heterogeneous group of myeloid malignancies associated with poor outcomes. Studies carried out in the last years have in part elucidated the complex role played by TP53 mutations in the pathogenesis of these myeloid disorders and in the mechanisms of drug resistance. A consistent number of studies has shown that some molecular parameters, such as the presence of a single or multiple TP53 mutations, the presence of concomitant TP53 deletions, the association with co-occurring mutations, the clonal size of TP53 mutations, the involvement of a single (monoallelic) or of both TP53 alleles (biallelic) and the cytogenetic architecture of concomitant chromosome abnormalities are major determinants of outcomes of patients. The limited response of these patients to standard treatments, including induction chemotherapy, hypomethylating agents and venetoclax-based therapies and the discovery of an immune dysregulation have induced a shift to new emerging therapies, some of which being associated with promising efficacy. The main aim of these novel immune and nonimmune strategies consists in improving survival and in increasing the number of TP53-mutated MDS/AML patients in remission amenable to allogeneic stem cell transplantation.
Collapse
Affiliation(s)
- Ugo Testa
- Department of Oncology, Istituto Superiore di Sanità, Rome Italy
| | - Germana Castelli
- Department of Oncology, Istituto Superiore di Sanità, Rome Italy
| | - Elvira Pelosi
- Department of Oncology, Istituto Superiore di Sanità, Rome Italy
| |
Collapse
|
20
|
Ziogas DC, Theocharopoulos C, Lialios PP, Foteinou D, Koumprentziotis IA, Xynos G, Gogas H. Beyond CTLA-4 and PD-1 Inhibition: Novel Immune Checkpoint Molecules for Melanoma Treatment. Cancers (Basel) 2023; 15:2718. [PMID: 37345056 PMCID: PMC10216291 DOI: 10.3390/cancers15102718] [Citation(s) in RCA: 36] [Impact Index Per Article: 18.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2023] [Revised: 05/04/2023] [Accepted: 05/08/2023] [Indexed: 06/23/2023] Open
Abstract
More than ten years after the approval of ipilimumab, immune checkpoint inhibitors (ICIs) against PD-1 and CTLA-4 have been established as the most effective treatment for locally advanced or metastatic melanoma, achieving durable responses either as monotherapies or in combinatorial regimens. However, a considerable proportion of patients do not respond or experience early relapse, due to multiple parameters that contribute to melanoma resistance. The expression of other immune checkpoints beyond the PD-1 and CTLA-4 molecules remains a major mechanism of immune evasion. The recent approval of anti-LAG-3 ICI, relatlimab, in combination with nivolumab for metastatic disease, has capitalized on the extensive research in the field and has highlighted the potential for further improvement of melanoma prognosis by synergistically blocking additional immune targets with new ICI-doublets, antibody-drug conjugates, or other novel modalities. Herein, we provide a comprehensive overview of presently published immune checkpoint molecules, including LAG-3, TIGIT, TIM-3, VISTA, IDO1/IDO2/TDO, CD27/CD70, CD39/73, HVEM/BTLA/CD160 and B7-H3. Beginning from their immunomodulatory properties as co-inhibitory or co-stimulatory receptors, we present all therapeutic modalities targeting these molecules that have been tested in melanoma treatment either in preclinical or clinical settings. Better understanding of the checkpoint-mediated crosstalk between melanoma and immune effector cells is essential for generating more effective strategies with augmented immune response.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Helen Gogas
- First Department of Medicine, Laiko General Hospital, School of Medicine, National and Kapodistrian University of Athens, 11527 Athens, Greece; (C.T.); (P.-P.L.); (D.F.); (I.-A.K.); (G.X.)
| |
Collapse
|
21
|
Liu SY, Mulugeta N, Dougan SK, Qiang L. In vitro flow cytometry assay to assess primary human and mouse macrophage phagocytosis of live cells. STAR Protoc 2023; 4:102240. [PMID: 37074910 PMCID: PMC10148074 DOI: 10.1016/j.xpro.2023.102240] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2022] [Revised: 03/06/2023] [Accepted: 03/23/2023] [Indexed: 04/20/2023] Open
Abstract
Although tumor-associated macrophages are generally immunosuppressive, macrophages may also promote tumor clearance via phagocytosis of live tumor cells. Here, we present a protocol for assessing macrophage engulfment of tumor cells in vitro using flow cytometry. We describe steps for cell preparation, reseeding macrophages, and setting up phagocytosis. We then detail procedures for collecting samples, staining macrophages, and flow cytometry. The protocol is applicable to both mouse bone-marrow-derived macrophages and human monocyte-derived macrophages. For complete details on the use and execution of this protocol, please refer to Roehle et al. (2021).1.
Collapse
Affiliation(s)
| | - Naomi Mulugeta
- Dana-Farber Cancer Institute, Boston, MA 02215, USA; Harvard College, Cambridge, MA 02138, USA
| | - Stephanie K Dougan
- Dana-Farber Cancer Institute, Boston, MA 02215, USA; Harvard Medical School, Boston, MA 02215, USA.
| | - Li Qiang
- Dana-Farber Cancer Institute, Boston, MA 02215, USA; Harvard Medical School, Boston, MA 02215, USA.
| |
Collapse
|
22
|
Ilan U, Brivio E, Algeri M, Balduzzi A, Gonzalez-Vincent M, Locatelli F, Zwaan CM, Baruchel A, Lindemans C, Bautista F. The Development of New Agents for Post-Hematopoietic Stem Cell Transplantation Non-Infectious Complications in Children. J Clin Med 2023; 12:2149. [PMID: 36983151 PMCID: PMC10054172 DOI: 10.3390/jcm12062149] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2023] [Revised: 02/27/2023] [Accepted: 03/01/2023] [Indexed: 03/12/2023] Open
Abstract
Hematopoietic stem cell transplantation (HSCT) is often the only curative treatment option for patients suffering from various types of malignant diseases and some non-cancerous conditions. Nevertheless, it is associated with a high risk of complications leading to transplant-related mortality and long-term morbidity. An increasing number of therapeutic and prevention strategies have been developed over the last few years to tackle the complications arising in patients receiving an HSCT. These strategies have been mainly carried out in adults and some are now being translated into children. In this manuscript, we review the recent advancements in the development and implementation of treatment options for post-HSCT non-infectious complications in pediatric patients with leukemia and other non-malignant conditions, with a special attention on the new agents available within clinical trials. We focused on the following conditions: graft failure, prevention of relapse and early interventions after detection of minimal residual disease positivity following HSCT in acute lymphoblastic and myeloid leukemia, chronic graft versus host disease, non-infectious pulmonary complications, and complications of endothelial origin.
Collapse
Affiliation(s)
- Uri Ilan
- Princess Máxima Center for Pediatric Oncology, 3584 CS Utrecht, The Netherlands
| | - Erica Brivio
- Princess Máxima Center for Pediatric Oncology, 3584 CS Utrecht, The Netherlands
| | - Mattia Algeri
- Department of Hematology/Oncology and Cell and Gene Therapy, Bambino Gesù Children Hospital, 00165 Rome, Italy
| | - Adriana Balduzzi
- Clinica Pediatrica Università degli Studi di Milano Bicocca, 20900 Monza, Italy
| | - Marta Gonzalez-Vincent
- Department of Stem Cell Transplantation, Hospital Infantil Universitario Nino Jesus, 28009 Madrid, Spain
| | - Franco Locatelli
- Department of Hematology/Oncology and Cell and Gene Therapy, Bambino Gesù Children Hospital, 00165 Rome, Italy
| | | | - Andre Baruchel
- Department of Pediatric Hematology, AP-HP, Robert Debré Hospital, 75019 Paris, France
| | - Caroline Lindemans
- Princess Máxima Center for Pediatric Oncology, 3584 CS Utrecht, The Netherlands
- Division of Pediatrics, University Medical Center Utrecht, 3584 CX Utrecht, The Netherlands
- Department of Stem Cell Transplantation, Regenerative Medicine Center, University Medical Center, 3584 CX Utrecht, The Netherlands
| | - Francisco Bautista
- Princess Máxima Center for Pediatric Oncology, 3584 CS Utrecht, The Netherlands
| |
Collapse
|
23
|
Zeidan AM, Giagounidis A, Sekeres MA, Xiao Z, Sanz GF, Hoef MV, Ma F, Hertle S, Santini V. STIMULUS-MDS2 design and rationale: a phase III trial with the anti-TIM-3 sabatolimab (MBG453) + azacitidine in higher risk MDS and CMML-2. Future Oncol 2023; 19:631-642. [PMID: 37083373 DOI: 10.2217/fon-2022-1237] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/22/2023] Open
Abstract
Patients with higher-risk myelodysplastic syndromes (MDS) and chronic myelomonocytic leukemia (CMML) unfit for hematopoietic stem cell transplantation have poor outcomes. Novel therapies that provide durable benefit with favorable tolerability and clinically meaningful improvement in survival are needed. T-cell immunoglobulin domain and mucin domain-3 (TIM-3) is an immuno-myeloid regulator expressed on immune and leukemic stem cells in myeloid malignancies. Sabatolimab is a novel immunotherapy targeting TIM-3 with a potential dual mechanism of reactivating the immune system and directly targeting TIM-3+ leukemic blasts suppressing the growth of cancer cells. Here, we describe the aims and design of the phase III STIMULUS-MDS2 trial, which aims to demonstrate the potential for sabatolimab plus azacitidine to improve survival for patients with higher-risk MDS and CMML-2 (NCT04266301). Clinical Trial Registration: NCT04266301 (ClinicalTrials.gov).
Collapse
Affiliation(s)
- Amer M Zeidan
- Yale University & Yale Cancer Center, New Haven, CT 06510, USA
| | | | - Mikkael A Sekeres
- Division of Hematology, Sylvester Cancer Center, University of Miami, Miami, FL 33065, USA
| | - Zhijian Xiao
- Blood Diseases Hospital, Chinese Academy of Medical Sciences, Tianjin, 300020, China
| | - Guillermo F Sanz
- Hospital Universitario y Politécnico La Fe, Valencia, 46026, Spain
- Health Research Institute La Fe (IIS La Fe), Valencia, 46026, Spain
- CIBERONC, Instituto de Salud Carlos III, Madrid, 28029, Spain
| | | | - Fei Ma
- Novartis Pharmaceuticals Corporation, East Hanover, NJ 07936, USA
| | | | - Valeria Santini
- MDS Unit, Hematology, University of Florence, Florence, 50121, Italy
| |
Collapse
|
24
|
Bailly C, Thuru X, Goossens L, Goossens JF. Soluble TIM-3 as a biomarker of progression and therapeutic response in cancers and other of human diseases. Biochem Pharmacol 2023; 209:115445. [PMID: 36739094 DOI: 10.1016/j.bcp.2023.115445] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2022] [Revised: 01/26/2023] [Accepted: 01/27/2023] [Indexed: 02/05/2023]
Abstract
Immune checkpoints inhibition is a privileged approach to combat cancers and other human diseases. The TIM-3 (T cell immunoglobulin and mucin-domain containing-3) inhibitory checkpoint expressed on different types of immune cells is actively investigated as an anticancer target, with a dozen of monoclonal antibodies in (pre)clinical development. A soluble form sTIM-3 can be found in the plasma of patients with cancer and other diseases. This active circulating protein originates from the proteolytic cleavage by two ADAM metalloproteases of the membrane receptor shared by tumor and non-tumor cells, and extracellular vesicles. In most cancers but not all, overexpression of mTIM-3 at the cell surface leads to high level of sTIM-3. Similarly, elevated levels of sTIM-3 have been reported in chronic autoimmune diseases, inflammatory gastro-intestinal diseases, certain viral and parasitic diseases, but also in cases of organ transplantation and in pregnancy-related pathologies. We have analyzed the origin of sTIM-3, its methods of dosage in blood or plasma, its presence in multiple diseases and its potential role as a biomarker to follow disease progression and/or the treatment response. In contrast to sPD-L1 generated by different classes of proteases and by alternative splicing, sTIM-3 is uniquely produced upon ADAM-dependent shedding, providing a more homogenous molecular entity and a possibly more reliable molecular marker. However, the biological functionality of sTIM-3 remains insufficiently characterized. The review shed light on pathologies associated with an altered expression of sTIM-3 in human plasma and the possibility to use sTIM-3 as a diagnostic or therapeutic marker.
Collapse
Affiliation(s)
- Christian Bailly
- OncoWitan, Consulting Scientific Office, Lille (Wasquehal) 59290, France; University of Lille, Faculty of Pharmacy, Institut de Chimie Pharmaceutique Albert Lespagnol (ICPAL), 3 rue du Professeur Laguesse, 59000 Lille, France; University of Lille, CNRS, Inserm, CHU Lille, UMR9020 - UMR1277 - Canther - Cancer Heterogeneity, Plasticity and Resistance to Therapies, 59000 Lille, France.
| | - Xavier Thuru
- University of Lille, CNRS, Inserm, CHU Lille, UMR9020 - UMR1277 - Canther - Cancer Heterogeneity, Plasticity and Resistance to Therapies, 59000 Lille, France
| | - Laurence Goossens
- University of Lille, Faculty of Pharmacy, Institut de Chimie Pharmaceutique Albert Lespagnol (ICPAL), 3 rue du Professeur Laguesse, 59000 Lille, France; University of Lille, CHU Lille, ULR 7365 - GRITA - Groupe de Recherche sur les formes Injectables et les Technologies Associées, 59000 Lille, France
| | - Jean-François Goossens
- University of Lille, CHU Lille, ULR 7365 - GRITA - Groupe de Recherche sur les formes Injectables et les Technologies Associées, 59000 Lille, France
| |
Collapse
|
25
|
Abstract
PURPOSE OF REVIEW Therapies that target the immune system are increasingly used across oncology, including in hematologic malignancies such as myelodysplastic syndromes (MDS) and acute myeloid leukemia (AML). While allogeneic transplant has been a key therapy in these cancers, new approaches that target the immune system are being explored including immune checkpoint therapies, antibody-drug conjugates, and cellular therapies. RECENT FINDINGS This review outlines updates in the preclinical rationale for immune directed therapies in MDS and AML, as well as recent clinical trials exploring these therapies. This manuscript summarizes the development of therapies targeting T cell immunoglobulin and mucin domain-containing protein 3 (TIM-3) and CD47, which are being evaluated in late phase studies in MDS and AML. It also reviews the landscape of other immune based therapies including antibody-drug conjugates, chimeric antigen receptor-T cells, bispecific antibodies, and tumor vaccines. SUMMARY The treatment landscape in MDS and AML is rapidly changing; with a goal of improving the quality and duration of responses, a number of immune based therapies are under investigation. This review outlines recent advances with these therapies as well as some of the challenges that remain to incorporate them into leukemia care.
Collapse
Affiliation(s)
- Andrew M Brunner
- Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts, USA
| |
Collapse
|
26
|
Venugopal S, Xie Z, Zeidan AM. An overview of novel therapies in advanced clinical testing for acute myeloid leukemia. Expert Rev Hematol 2023; 16:109-119. [PMID: 36718500 DOI: 10.1080/17474086.2023.2174521] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/01/2023]
Abstract
INTRODUCTION The past decade has seen a sea change in the AML landscape with vastly improved cognizance of molecular pathogenesis, clonal evolution, and importance of measurable residual disease. Since 2017, the therapeutic armamentarium of AML has considerably expanded with the approval of midostaurin, enasidenib, ivosidenib, gilteritinib, and venetoclax in combination with hypomethylating agents and others. Nevertheless, relapse and treatment refractoriness remain the insurmountable challenges in AML therapy. This has galvanized the leukemic research community leading to the discovery and development of agents that specifically target gene mutations, molecularly agnostic therapies that exploit immune environment, apoptotic pathways, leukemic cell surface antigens and so forth. AREAS COVERED This article provides an overview of the pathophysiology of AML in the context of non-cellular immune and molecularly targeted and agnostic therapies that are in clinical trial development in AML. EXPERT OPINION Ever growing understanding of the molecular pathogenesis and metabolomics in AML has allowed the researchers to identify targets directed at specific genes and metabolic pathways. As a result, AML therapy is constantly evolving and so are the escape mechanisms leading to disease relapse. Therefore, it is of paramount importance to sequentially evaluate the patient during AML treatment and intervene at the right time.
Collapse
Affiliation(s)
- Sangeetha Venugopal
- Division of Hematology, Sylvester Comprehensive Cancer Center, University of Miami, Miami, FL, USA
| | - Zhuoer Xie
- Department of Malignant Hematology, H. Lee Moffitt Cancer Center & Research Institute, FL, USA
| | - Amer M Zeidan
- Section of Hematology, Department of Internal Medicine, Yale University School of Medicine, New Haven, CT, USA
| |
Collapse
|
27
|
Bailly C. Contribution of the TIM-3/Gal-9 immune checkpoint to tropical parasitic diseases. Acta Trop 2023; 238:106792. [PMID: 36509129 DOI: 10.1016/j.actatropica.2022.106792] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2022] [Revised: 11/23/2022] [Accepted: 12/05/2022] [Indexed: 12/13/2022]
Abstract
Neglected tropical parasitic diseases (NTD) are prevalent in many countries and cost-effective treatments remain urgently needed. Novel approaches have been proposed to address these diseases through an action on immune co-inhibitory checkpoints which are exploited by parasites to evade the immune system. Among these checkpoints, TIM-3 has been shown to play a key role in antiparasitic immunity via a repression and functional attenuation of CD4+ and/or CD8+ T-cells. The present review discusses the role of the TIM-3/galectin-9 checkpoint in seven major NTD: Chagas disease, leishmaniasis and malaria (3 trypanosomatid infections), schistosomiasis, toxoplasmosis, echinococcosis and filariasis (4 helminth infections). In each case, the role of the checkpoint has been analyzed and the use of anti-TIM-3 antibodies evaluated as a potential therapeutic approach. In general, the parasitic infection is coupled with an upregulation of TIM-3 expressed on T cells, but not necessarily with an exhaustion of those T cells. In several cases, the use of anti-TIM-3 antibodies represent a possible strategy to reinforce the clearance and to reduce the parasite load. Promising data have been reported in cases of leishmaniasis, malaria and schistosomiasis, whereas a similar approach proved much less efficient (if not deleterious) in cases of echinococcosis and the Chagas disease. Nevertheless, the TIM-3 checkpoint warrants further consideration as a potential immune target to combat these pathologies, using antibodies or drugs capable of reducing directly or indirectly the expression and function of the checkpoint, to restore an immune control.
Collapse
Affiliation(s)
- Christian Bailly
- OncoWitan, Scientific Consulting Office, Lille (Wasquehal), 59290, France; University of Lille, Institut de Chimie Pharmaceutique Albert Lespagnol (ICPAL), Faculté de Pharmacie, 3 rue du Professeur Laguesse, BP-83, F-59006, Lille, France.
| |
Collapse
|
28
|
Morse JW, Rios M, Ye J, Rios A, Zhang CC, Daver NG, DiNardo CD, Zhang N, An Z. Antibody therapies for the treatment of acute myeloid leukemia: exploring current and emerging therapeutic targets. Expert Opin Investig Drugs 2023; 32:107-125. [PMID: 36762937 PMCID: PMC10031751 DOI: 10.1080/13543784.2023.2179482] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2022] [Accepted: 02/08/2023] [Indexed: 02/11/2023]
Abstract
INTRODUCTION Acute myeloid leukemia (AML) is the most common and deadly type of leukemia affecting adults. It is typically managed with rounds of non-targeted chemotherapy followed by hematopoietic stem cell transplants, but this is only possible in patients who can tolerate these harsh treatments and many are elderly and frail. With the identification of novel tumor-specific cell surface receptors, there is great conviction that targeted antibody therapies will soon become available for these patients. AREAS COVERED In this review, we describe the current landscape of known target receptors for monospecific and bispecific antibody-based therapeutics for AML. Here, we characterize each of the receptors and targeted antibody-based therapeutics in development, illustrating the rational design behind each therapeutic compound. We then discuss the bispecific antibodies in development and how they improve immune surveillance of AML. For each therapeutic, we also summarize the available pre-clinical and clinical data, including data from discontinued trials. EXPERT OPINION One antibody-based therapeutic has already been approved for AML treatment, the CD33-targeting antibody-drug conjugate, gemtuzumab ozogamicin. Many more are currently in pre-clinical and clinical studies. These antibody-based therapeutics can perform tumor-specific, elaborate cytotoxic functions and there is growing confidence they will soon lead to personalized, safe AML treatment options that induce durable remissions.
Collapse
Affiliation(s)
- Joshua W Morse
- Texas Therapeutics Institute, Brown Foundation Institute of Molecular Medicine, The University of Texas Health Science Center at Houston, Houston, Texas, USA
| | - Margarita Rios
- Gorgas Memorial Institute of Health Studies, Panama City, Panama
| | - John Ye
- Texas Therapeutics Institute, Brown Foundation Institute of Molecular Medicine, The University of Texas Health Science Center at Houston, Houston, Texas, USA
| | - Adan Rios
- Division of Oncology, Department of Internal Medicine, The University of Texas Health Science Center at Houston, Houston, Texas, USA
| | - Cheng Cheng Zhang
- Department of Physiology, University of Texas Southwestern Medical Center, Dallas, Texas, USA
| | - Naval G Daver
- Department of Leukemia, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Courtney D DiNardo
- Department of Leukemia, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Ningyan Zhang
- Texas Therapeutics Institute, Brown Foundation Institute of Molecular Medicine, The University of Texas Health Science Center at Houston, Houston, Texas, USA
| | - Zhiqiang An
- Texas Therapeutics Institute, Brown Foundation Institute of Molecular Medicine, The University of Texas Health Science Center at Houston, Houston, Texas, USA
| |
Collapse
|
29
|
Barth S, Naran K. TIM-3: a tumor-associated antigen beyond checkpoint inhibition? IMMUNOTHERAPY ADVANCES 2022; 2:ltac021. [PMID: 36406467 PMCID: PMC9669666 DOI: 10.1093/immadv/ltac021] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2022] [Accepted: 10/21/2022] [Indexed: 11/18/2022] Open
Abstract
Immune checkpoint inhibitors are one of the most remarkable immunomodulatory therapies of current times. Sabatolimab is a high-affinity, humanized anti-TIM-3 monoclonal antibody currently in development for patients with myeloproliferative disorders, including acute myeloid leukemia and myelodysplastic syndromes. By targeting TIM-3, a receptor expressed on various immune effector cells as well as myeloid cells, multiple mechanisms of action that are distinct from canonical immune checkpoint inhibitors are in play - (i) blockade of TIM-3 and its ligands PtdSer/galectin-9, (ii) modulation of leukemic cell self-renewal as well as (iii) antibody-dependent phagocytosis of TIM-3-expressing leukemic cells. Novel immunotherapies such as sabatolimab which enhance the antitumor immune response on converging fronts represent the promise of a continuously replenished armoury for the treatment of cancer.
Collapse
Affiliation(s)
- Stefan Barth
- Medical Biotechnology and Immunotherapy Research Unit, Institute of Infectious Disease and Molecular Medicine, Faculty of Health Sciences, University of Cape Town, Cape Town, South Africa
- South African Research Chair in Cancer Biotechnology, Department of Integrative Biomedical Sciences, Faculty of Health Sciences, University of Cape Town, Cape Town, South Africa
| | - Krupa Naran
- Medical Biotechnology and Immunotherapy Research Unit, Institute of Infectious Disease and Molecular Medicine, Faculty of Health Sciences, University of Cape Town, Cape Town, South Africa
| |
Collapse
|