1
|
Sampson JF, Zhang H, Zhang D, Bi M, Hinthorne A, Syed S, Zhang Y, Chattopadhyay N, Collins S, Pogue S, Björck P, Curley M. CD38-targeted attenuated interferon alpha immunocytokine activates both innate and adaptive immune cells to drive anti-tumor activity. PLoS One 2025; 20:e0321622. [PMID: 40315226 PMCID: PMC12047799 DOI: 10.1371/journal.pone.0321622] [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: 07/25/2024] [Accepted: 03/10/2025] [Indexed: 05/04/2025] Open
Abstract
Recombinant interferon alpha (IFNα) has been used to treat cancer patients for over 30 years; however, its clinical utility has been limited by a narrow therapeutic index. Given the recognized anti-tumor and immunomodulatory impacts of IFNα, the development of novel strategies to harness these attributes while minimizing associated toxicity could provide significant benefit for patients. The concept of attenuating IFNα binding affinity for its receptor was conceived to address this challenge and led to the development of CD38-targeted Attenukine™, a CD38-targeted antibody attenuated IFNα immunocytokine. In this study, we sought to delineate the effects of targeting AttenukineTM specifically to tumor cells and/or immune cells using an antibody to CD38, a cell surface glycoprotein expressed on certain tumor and immune cells, using different mouse models and anti-human or anti-mouse CD38-targeted Attenukine™. Our results demonstrate that an anti-human CD38 AttenukineTM inhibits tumor growth through direct anti-proliferative effects of IFNα on CD38 + tumor cells as well as by indirectly modulating the anti-tumor immune response. In various in vivo models leveraging syngeneic mice bearing tumors with or without CD38 expression, administration of CD38-murine AttenukineTM mediated anti-tumor efficacy with increased immune activation and intra-tumoral infiltration. These data point to a potential dual mechanism of action for CD38-targeted Attenukine™, involving both tumor- and immune-directed effects, and highlight the potential benefit of a CD38-targeted attenuated IFNα therapy to deliver the known effects of IFNαtreatment to a broad spectrum of patients, while limiting the toxicity typically associated with recombinant IFNα.
Collapse
Affiliation(s)
- James F. Sampson
- Oncology Drug Discovery Unit, Takeda Development Center Americas, Inc. (TDCA), Lexington, Massachusetts, United States of America
| | - Hong Zhang
- Oncology Drug Discovery Unit, Takeda Development Center Americas, Inc. (TDCA), Lexington, Massachusetts, United States of America
| | - Dongmei Zhang
- Oncology Drug Discovery Unit, Takeda Development Center Americas, Inc. (TDCA), Lexington, Massachusetts, United States of America
| | - Mingying Bi
- Discovery R and D, Teva Pharmaceutical Industries, Ltd., Redwood City, United States of America
| | - Adam Hinthorne
- Oncology Drug Discovery Unit, Takeda Development Center Americas, Inc. (TDCA), Lexington, Massachusetts, United States of America
| | - Sakeena Syed
- Oncology Drug Discovery Unit, Takeda Development Center Americas, Inc. (TDCA), Lexington, Massachusetts, United States of America
| | - Yuhong Zhang
- Oncology Drug Discovery Unit, Takeda Development Center Americas, Inc. (TDCA), Lexington, Massachusetts, United States of America
| | - Nibedita Chattopadhyay
- Oncology Drug Discovery Unit, Takeda Development Center Americas, Inc. (TDCA), Lexington, Massachusetts, United States of America
| | - Sabrina Collins
- Oncology Drug Discovery Unit, Takeda Development Center Americas, Inc. (TDCA), Lexington, Massachusetts, United States of America
| | - Sarah Pogue
- Discovery R and D, Teva Pharmaceutical Industries, Ltd., Redwood City, United States of America
| | - Pia Björck
- Discovery R and D, Teva Pharmaceutical Industries, Ltd., Redwood City, United States of America
| | - Michael Curley
- Oncology Drug Discovery Unit, Takeda Development Center Americas, Inc. (TDCA), Lexington, Massachusetts, United States of America
| |
Collapse
|
2
|
Senapati J, Jabbour E. Dar-ting at CD38 in T-cell leukemias. Blood 2024; 144:2162-2164. [PMID: 39570597 DOI: 10.1182/blood.2024026118] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2024] Open
|
3
|
Xu R, He X, Xu J, Yu G, Wu Y. Immunometabolism: signaling pathways, homeostasis, and therapeutic targets. MedComm (Beijing) 2024; 5:e789. [PMID: 39492834 PMCID: PMC11531657 DOI: 10.1002/mco2.789] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2023] [Revised: 09/20/2024] [Accepted: 09/25/2024] [Indexed: 11/05/2024] Open
Abstract
Immunometabolism plays a central role in sustaining immune system functionality and preserving physiological homeostasis within the organism. During the differentiation and activation, immune cells undergo metabolic reprogramming mediated by complex signaling pathways. Immune cells maintain homeostasis and are influenced by metabolic microenvironmental cues. A series of immunometabolic enzymes modulate immune cell function by metabolizing nutrients and accumulating metabolic products. These enzymes reverse immune cells' differentiation, disrupt intracellular signaling pathways, and regulate immune responses, thereby influencing disease progression. The huge population of immune metabolic enzymes, the ubiquity, and the complexity of metabolic regulation have kept the immune metabolic mechanisms related to many diseases from being discovered, and what has been revealed so far is only the tip of the iceberg. This review comprehensively summarized the immune metabolic enzymes' role in multiple immune cells such as T cells, macrophages, natural killer cells, and dendritic cells. By classifying and dissecting the immunometabolism mechanisms and the implications in diseases, summarizing and analyzing advancements in research and clinical applications of the inhibitors targeting these enzymes, this review is intended to provide a new perspective concerning immune metabolic enzymes for understanding the immune system, and offer novel insight into future therapeutic interventions.
Collapse
Affiliation(s)
- Rongrong Xu
- National Key Laboratory of Immunity and Inflammation & Institute of ImmunologyCollege of Basic Medical SciencesNaval Medical UniversityShanghaiChina
- School of Life SciencesFudan UniversityShanghaiChina
| | - Xiaobo He
- National Key Laboratory of Immunity and Inflammation & Institute of ImmunologyCollege of Basic Medical SciencesNaval Medical UniversityShanghaiChina
| | - Jia Xu
- National Key Laboratory of Immunity and Inflammation & Institute of ImmunologyCollege of Basic Medical SciencesNaval Medical UniversityShanghaiChina
| | - Ganjun Yu
- National Key Laboratory of Immunity and Inflammation & Institute of ImmunologyCollege of Basic Medical SciencesNaval Medical UniversityShanghaiChina
| | - Yanfeng Wu
- National Key Laboratory of Immunity and Inflammation & Institute of ImmunologyCollege of Basic Medical SciencesNaval Medical UniversityShanghaiChina
| |
Collapse
|
4
|
Nair AG, Leon-Ponte M, Kim VHD, Sussman G, Ehrhardt GR, Grunebaum E. Characterizing CD38 expression in terminally differentiated B cells using variable lymphocyte receptor B tetramers. Front Immunol 2024; 15:1451232. [PMID: 39575239 PMCID: PMC11579616 DOI: 10.3389/fimmu.2024.1451232] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2024] [Accepted: 09/30/2024] [Indexed: 11/24/2024] Open
Abstract
Introduction CD38 is an ectoenzyme receptor found on hematopoietic cells and its expression is used in the flow cytometric analysis of sub-populations of circulating B cells among peripheral blood mononuclear cells (PBMC) to aid in diagnosing patients with different antibody production defects (AbD). Monoclonal antibodies derived from the sea lamprey Variable Lymphocyte Receptor B (VLRB) are emerging as an alternative to conventional mammalian antibodies. We hypothesized that VLRB MM3 (V-CD38) which specifically recognizes CD38 in a manner correlating with its enzymatic activity could identify terminally differentiated B cells in human PBMC. Here we investigate the ability of V-CD38 as a tool to diagnose patients with diverse immune abnormalities including AbD. Methods The expression of CD38 on CD3-CD19+CD27+ plasmablasts and CD3-CD19+IgMhiCD27- transitional B cells in PBMC was analyzed by flow cytometry using V-CD38 and compared with a commercial conventional antibody to CD38 (C-CD38). Results A highly significant correlation (p<0.001, r=0.99) between the percentages of plasmablasts recognized by V-CD38 and C-CD38 was observed among 36 healthy controls (HC), 7 patients with AbD and 24 allergic individuals (AI). The use of V-CD38 enabled improved gating of the CD38 expressing cells (CD38+), aiding in the observation that patients with AbD had significantly lower (p=0.002) CD38+ plasmablasts (0.13%±0.13%) than HC (0.52%±0.57%). Only 61.3% of the transitional B cells detected by C-CD38 were also recognized by V-CD38 (r=0.95, p<0.001) among the 67 participants. AI had significantly reduced V-CD38 and C-CD38 transitional cells compared to HC (p=0.026 and p=0.012, respectively). Conclusions V-CD38 is a novel reagent that can assess B cells in human PBMC.
Collapse
Affiliation(s)
- Arundhati G. Nair
- Developmental and Stem Cell Biology Program, Hospital for Sick Children, Toronto, ON, Canada
- Institute of Medical Sciences, University of Toronto, Toronto, ON, Canada
| | - Matilde Leon-Ponte
- Developmental and Stem Cell Biology Program, Hospital for Sick Children, Toronto, ON, Canada
| | - Vy HD Kim
- Division of Immunology and Allergy, Department of Pediatrics, The Hospital for Sick Children, Toronto, ON, Canada
| | - Gordon Sussman
- Division of Clinical Immunology and Allergy, Department of Medicine, University of Toronto, Toronto, ON, Canada
| | | | - Eyal Grunebaum
- Developmental and Stem Cell Biology Program, Hospital for Sick Children, Toronto, ON, Canada
- Institute of Medical Sciences, University of Toronto, Toronto, ON, Canada
- Division of Immunology and Allergy, Department of Pediatrics, The Hospital for Sick Children, Toronto, ON, Canada
- Department of Immunology, University of Toronto, Toronto, ON, Canada
| |
Collapse
|
5
|
Singh A, Patil J, Ghogale SG, Deshpande N, Girase K, Shetye N, Rajpal S, Chatterjee G, Patkar N, Jain D, Epari S, Shet T, Gujral S, Subramanian PG, Tembhare PR. Utility of leukocyte-associated immunoglobulin-like receptor-1 (CD305) in flow cytometric detection of minimal bone marrow involvement by B-cell non-Hodgkin lymphoma. CYTOMETRY. PART B, CLINICAL CYTOMETRY 2024; 106:359-369. [PMID: 39031805 DOI: 10.1002/cyto.b.22193] [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: 12/29/2023] [Revised: 05/29/2024] [Accepted: 06/10/2024] [Indexed: 07/22/2024]
Abstract
Multicolor flow cytometry (MFC) is crucial in detecting occult or minimal bone marrow (BM) involvement by non-Hodgkin lymphomas (NHL), which may not be detected using trephine biopsy or imaging studies. Detection of low-level BM involvement can be challenging without definite immunophenotypic aberrancies. We studied the utility of CD305 in MFC detection of minimal BM involvement by B-NHL, especially in the absence of aberrancies by commonly used markers. The study included 1084 consecutive BM samples submitted for the staging of B-NHLs (excluding CLL) over two years. Samples were studied for morphological, immunophenotypic, and histopathological assessment. MFC studies were performed using 10-13 color MFC, including CD305-antibody (clone, DX26). Minimal BM involvement was defined with a cutoff of ≤10% lymphoma cells in viable cells on MFC assessment. Of 1084, 148 samples revealed overt morphological involvement by B-NHL and were excluded from analysis. BM samples of 172/936 patients were morphologically negative but revealed involvement using MFC independently. Corresponding trephine biopsy involvement was detected in only 79/172 (45.9%) patients. On MFC, 23/172 samples showed BM involvement with >10% lymphoma cells, and 149/172 (86.6%) samples revealed minimal involvement. In 54/149 (36.24%) samples, lymphoma cells were detected only with aberrant loss of CD305 expression. In 78 of the remaining 95 samples (82.1%), it provided an immunophenotypic aberrancy addition to other markers and supported the results. CD305 is a highly useful marker in the flow cytometric assessment of minimal BM involvement by B-NHL. MFC is a superior modality to trephine biopsy in detecting low-level BM involvement.
Collapse
Affiliation(s)
- Anu Singh
- Hematopathology Department, ACTREC, Tata Memorial Center, Homi Bhabha National Institute (HBNI) University, Navi Mumbai, Maharashtra, India
| | - Jagruti Patil
- Hematopathology Department, ACTREC, Tata Memorial Center, Homi Bhabha National Institute (HBNI) University, Navi Mumbai, Maharashtra, India
| | - Sitaram G Ghogale
- Hematopathology Department, ACTREC, Tata Memorial Center, Homi Bhabha National Institute (HBNI) University, Navi Mumbai, Maharashtra, India
| | - Nilesh Deshpande
- Hematopathology Department, ACTREC, Tata Memorial Center, Homi Bhabha National Institute (HBNI) University, Navi Mumbai, Maharashtra, India
| | - Karishma Girase
- Hematopathology Department, ACTREC, Tata Memorial Center, Homi Bhabha National Institute (HBNI) University, Navi Mumbai, Maharashtra, India
| | - Navami Shetye
- Hematopathology Department, ACTREC, Tata Memorial Center, Homi Bhabha National Institute (HBNI) University, Navi Mumbai, Maharashtra, India
| | - Sweta Rajpal
- Hematopathology Department, ACTREC, Tata Memorial Center, Homi Bhabha National Institute (HBNI) University, Navi Mumbai, Maharashtra, India
| | - Gaurav Chatterjee
- Hematopathology Department, ACTREC, Tata Memorial Center, Homi Bhabha National Institute (HBNI) University, Navi Mumbai, Maharashtra, India
| | - Nikhil Patkar
- Hematopathology Department, ACTREC, Tata Memorial Center, Homi Bhabha National Institute (HBNI) University, Navi Mumbai, Maharashtra, India
| | - Disha Jain
- Hematopathology Department, ACTREC, Tata Memorial Center, Homi Bhabha National Institute (HBNI) University, Navi Mumbai, Maharashtra, India
| | - Sridhar Epari
- Department of Pathology, Tata Memorial Center, HBNI University, Mumbai, Maharashtra, India
| | - Tanuja Shet
- Department of Pathology, Tata Memorial Center, HBNI University, Mumbai, Maharashtra, India
| | - Sumeet Gujral
- Hematopathology Department, ACTREC, Tata Memorial Center, Homi Bhabha National Institute (HBNI) University, Navi Mumbai, Maharashtra, India
- Department of Pathology, Tata Memorial Center, HBNI University, Mumbai, Maharashtra, India
| | - Papagudi G Subramanian
- Hematopathology Department, ACTREC, Tata Memorial Center, Homi Bhabha National Institute (HBNI) University, Navi Mumbai, Maharashtra, India
| | - Prashant R Tembhare
- Hematopathology Department, ACTREC, Tata Memorial Center, Homi Bhabha National Institute (HBNI) University, Navi Mumbai, Maharashtra, India
| |
Collapse
|
6
|
Shi H, Yang F, Cao M, Xu T, Zheng P, Guo Y, Su G, Feng S, Li R, Liu R, Liu H, Ma L, Ke X, Hu K. Daratumumab and venetoclax combined with CAGE for late R/R T-ALL/LBL patients: Single-arm, open-label, phase I study. Ann Hematol 2024; 103:2993-3004. [PMID: 38662205 DOI: 10.1007/s00277-024-05775-z] [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: 01/29/2024] [Accepted: 04/22/2024] [Indexed: 04/26/2024]
Abstract
The prognosis of patients diagnosed with relapsed or refractory (R/R) T-lymphoblastic leukemia/lymphoma (T-ALL/LBL) has consistently been unsatisfactory, with limited treatment options. As reports, the CAG regimen can serve as a salvage treatment for R/R T-ALL/LBL, but there remains a subset of patients who do not benefit from it. Recent studies have indicated that daratumumab (Dara) and venetoclax (Ven) may offer promising therapeutic benefits for T-ALL/LBL. In light of these findings, we conducted a safety and efficacy evaluation of the enhanced treatment regimen, combining Dara and Ven with aclarubicin, cytarabine, granulocyte colony-stimulating factor, and etoposide (CAGE), in patients suffering from R/R T-ALL/LBL. The participants in this phase I trial were patients with R/R T-ALL/LBL who fail to standard treatment regimens. During each 28-day cycle, the patients were treated by Dara, Ven, cytarabine, aclarubicin, granulocyte colony-stimulating factor, etoposide. The primary endpoint of this study was the rate of remission. This report presents the prospective outcomes of 21 patients who received the salvage therapy of Dara and Ven combined with the CAGE regimen (Dara + Ven + CAGE). The objective remission rate (ORR) was determined to be 57.1%, while the complete remission (CR) rate was 47.6%. Notably, patients with the early T-cell precursor (ETP) subtype exhibited a significantly higher remission rate in the bone marrow compared to non-ETP patients (100% vs. 44.4%, p = 0.044). The Dara + Ven + CAGE regimen demonstrated a favorable remission rate in patients with R/R T-ALL/LBL. Moreover, the treatment was well-tolerated.
Collapse
Affiliation(s)
- Hui Shi
- Department of Lymphoma and Myeloma Research Center, Beijing Gobroad Boren Hospital, Beijing, 100070, China
| | - Fan Yang
- Department of Lymphoma and Myeloma Research Center, Beijing Gobroad Boren Hospital, Beijing, 100070, China
| | - Miaomiao Cao
- Department of Lymphoma and Myeloma Research Center, Beijing Gobroad Boren Hospital, Beijing, 100070, China
| | - Teng Xu
- Department of Lymphoma and Myeloma Research Center, Beijing Gobroad Boren Hospital, Beijing, 100070, China
| | - Peihao Zheng
- Department of Lymphoma and Myeloma Research Center, Beijing Gobroad Boren Hospital, Beijing, 100070, China
| | - Yuelu Guo
- Department of Lymphoma and Myeloma Research Center, Beijing Gobroad Boren Hospital, Beijing, 100070, China
| | - Guoai Su
- Department of Lymphoma and Myeloma Research Center, Beijing Gobroad Boren Hospital, Beijing, 100070, China
| | - Shaomei Feng
- Department of Lymphoma and Myeloma Research Center, Beijing Gobroad Boren Hospital, Beijing, 100070, China
| | - Ruiting Li
- Department of Lymphoma and Myeloma Research Center, Beijing Gobroad Boren Hospital, Beijing, 100070, China
| | - Rui Liu
- Department of Lymphoma and Myeloma Research Center, Beijing Gobroad Boren Hospital, Beijing, 100070, China
| | - Haidi Liu
- Department of Lymphoma and Myeloma Research Center, Beijing Gobroad Boren Hospital, Beijing, 100070, China
| | - Lixia Ma
- Department of Lymphoma and Myeloma Research Center, Beijing Gobroad Boren Hospital, Beijing, 100070, China
| | - Xiaoyan Ke
- Department of Lymphoma and Myeloma Research Center, Beijing Gobroad Boren Hospital, Beijing, 100070, China.
- Department of Hematology, Peking University Third Hospital, Beijing, 100191, China.
| | - Kai Hu
- Department of Lymphoma and Myeloma Research Center, Beijing Gobroad Boren Hospital, Beijing, 100070, China.
| |
Collapse
|
7
|
Oh BL, Vinanica N, Wong DM, Campana D. Chimeric antigen receptor T-cell therapy for T-cell acute lymphoblastic leukemia. Haematologica 2024; 109:1677-1688. [PMID: 38832423 PMCID: PMC11141683 DOI: 10.3324/haematol.2023.283848] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2024] [Accepted: 01/11/2024] [Indexed: 06/05/2024] Open
Abstract
Chimeric antigen receptor (CAR) T-cell therapy is a new and effective treatment for patients with hematologic malignancies. Clinical responses to CAR T cells in leukemia, lymphoma, and multiple myeloma have provided strong evidence of the antitumor activity of these cells. In patients with refractory or relapsed B-cell acute lymphoblastic leukemia (ALL), the infusion of autologous anti-CD19 CAR T cells is rapidly gaining standard-of-care status and might eventually be incorporated into frontline treatment. In T-ALL, however, leukemic cells generally lack surface molecules recognized by established CAR, such as CD19 and CD22. Such deficiency is particularly important, as outcome is dismal for patients with T-ALL that is refractory to standard chemotherapy and/or hematopoietic stem cell transplant. Recently, CAR T-cell technologies directed against T-cell malignancies have been developed and are beginning to be tested clinically. The main technical obstacles stem from the fact that malignant and normal T cells share most surface antigens. Therefore, CAR T cells directed against T-ALL targets might be susceptible to self-elimination during manufacturing and/or have suboptimal activity after infusion. Moreover, removing leukemic cells that might be present in the cell source used for CAR T-cell manufacturing might be problematic. Finally, reconstitution of T cells and natural killer cells after CAR T-cell infusion might be impaired. In this article, we discuss potential targets for CAR T-cell therapy of T-ALL with an emphasis on CD7, and review CAR configurations as well as early clinical results.
Collapse
Affiliation(s)
- Bernice L.Z. Oh
- Viva-University Children’s Cancer Center, Khoo Teck Puat-National University Children’s Medical Institute, National University Hospital, National University Health System
- Department of Pediatrics, Yong Loo Lin School of Medicine, National University of Singapore
| | - Natasha Vinanica
- Department of Pediatrics, Yong Loo Lin School of Medicine, National University of Singapore
| | - Desmond M.H. Wong
- Department of Pediatrics, Yong Loo Lin School of Medicine, National University of Singapore
| | - Dario Campana
- Department of Pediatrics, Yong Loo Lin School of Medicine, National University of Singapore
- Cancer Science Institute, Yong Loo Lin School of Medicine, National University of Singapore, Singapore
| |
Collapse
|
8
|
Koslowski S, Glauben R, Habringer S, Burmeister T, Keller U, Brüggemann M, Gökbuget N, Schwartz S. Frequent, high density expression of surface CD38 as a potential therapeutic target in adult T-lineage acute lymphoblastic leukemia. Haematologica 2024; 109:661-665. [PMID: 37675513 PMCID: PMC10828765 DOI: 10.3324/haematol.2023.283814] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2023] [Accepted: 09/01/2023] [Indexed: 09/08/2023] Open
Affiliation(s)
- Sebastian Koslowski
- Department of Hematology, Oncology and Cancer Immunology (Campus Benjamin Franklin), Charité - Universitätsmedizin Berlin, corporate member of Freie Universität and Humboldt-Universität zu Berlin, Berlin
| | - Rainer Glauben
- Department of Gastroenterology, Infectious Diseases and Rheumatology (Campus Benjamin Franklin), Charité - Universitätsmedizin Berlin, corporate member of Freie Universität and Humboldt-Universität zu Berlin, Berlin
| | - Stefan Habringer
- Department of Hematology, Oncology and Cancer Immunology (Campus Benjamin Franklin), Charité - Universitätsmedizin Berlin, corporate member of Freie Universität and Humboldt-Universität zu Berlin, Berlin, Germany; German Cancer Consortium (DKTK) and German Cancer Research Center (DKFZ), Heidelberg
| | - Thomas Burmeister
- Department of Hematology, Oncology and Cancer Immunology (Campus Virchow-Klinikum), Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin
| | - Ulrich Keller
- Department of Hematology, Oncology and Cancer Immunology (Campus Benjamin Franklin), Charité - Universitätsmedizin Berlin, corporate member of Freie Universität and Humboldt-Universität zu Berlin, Berlin, Germany; German Cancer Consortium (DKTK) and German Cancer Research Center (DKFZ), Heidelberg, Germany; Max-Delbrück-Center, Berlin
| | - Monika Brüggemann
- University Cancer Center Schleswig-Holstein (UCCSH), University Hospital Schleswig-Holstein, Kiel, Germany; Department of Medicine II, Hematology and Oncology, University Hospital Schleswig-Holstein, Kiel
| | - Nicola Gökbuget
- German Cancer Consortium (DKTK) and German Cancer Research Center (DKFZ), Heidelberg, Germany; Medizinische Klinik II, Universitätsklinikum der Johann-Wolfgang-Goethe Universität, Frankfurt/Main
| | - Stefan Schwartz
- Department of Hematology, Oncology and Cancer Immunology (Campus Benjamin Franklin), Charité - Universitätsmedizin Berlin, corporate member of Freie Universität and Humboldt-Universität zu Berlin, Berlin, Germany; German Cancer Consortium (DKTK) and German Cancer Research Center (DKFZ), Heidelberg.
| |
Collapse
|
9
|
Prejzner W, Piekoś O, Bełdzińska K, Sadowska-Klasa A, Zarzycka E, Bieniaszewska M, Lewandowski K, Zaucha JM. The role of daratumumab in relapsed/refractory CD38 positive acute leukemias-case report on three cases with a literature review. Front Oncol 2023; 13:1228481. [PMID: 37941558 PMCID: PMC10628456 DOI: 10.3389/fonc.2023.1228481] [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] [Received: 05/24/2023] [Accepted: 08/16/2023] [Indexed: 11/10/2023] Open
Abstract
Primary refractory or relapsed T-cell acute lymphoblastic leukemia (T-ALL) and mixed phenotype myeloid/T-cell acute leukemia have dismal prognoses. New treatment approaches, preferably targeting specific leukemic aberrations to overcome resistance, are urgently needed. The bright expression of the CD38 antigen found in several cases of T-ALL led to an investigation into the role of anti-CD38 antibodies in the treatment of T-ALL. Here, we present three cases of resistant and relapsed T-ALL and myeloid/T-cell treated with daratumumab-based therapy, including venetoclax and bortezomib (Dara-Ven-Bor). All patients achieved complete remission, with minimal residual disease negativity within four weeks of treatment, allowing them to proceed to allogeneic hematopoietic cell transplantation. The toxicity of the triple schema was acceptable. Our patients and other cases reviewed here suggest that daratumumab combined with venetoclax and bortezomib may be a very effective and relatively safe salvage treatment, even in primary resistant T-ALL.
Collapse
Affiliation(s)
- Witold Prejzner
- Department of Hematology and Transplantology, Medical University of Gdansk, Gdańsk, Poland
| | - Oliwia Piekoś
- Department of Hematology and Transplantology, Medical University of Gdansk, Gdańsk, Poland
| | - Karolina Bełdzińska
- Department of Hematology and Transplantology, Medical University of Gdansk, Gdańsk, Poland
| | - Alicja Sadowska-Klasa
- Department of Hematology and Transplantology, Medical University of Gdansk, Gdańsk, Poland
| | - Ewa Zarzycka
- Department of Hematology and Transplantology, Medical University of Gdansk, Gdańsk, Poland
| | - Maria Bieniaszewska
- Department of Hematology and Transplantology, Medical University of Gdansk, Gdańsk, Poland
| | | | - Jan Maciej Zaucha
- Department of Hematology and Transplantology, Medical University of Gdansk, Gdańsk, Poland
| |
Collapse
|
10
|
Saha A, Brahmbhatt B, Rai V, Kakoty S, Sawhney J. A Comparative Analysis of Early T-Cell Precursor Lymphoblastic Leukemia/Lymphoma (ETP-ALL/LBL) and Non-ETP-ALL/LBL in a Tertiary Cancer Care Center Based in Western India. Indian J Hematol Blood Transfus 2023; 39:699-704. [PMID: 37786816 PMCID: PMC10542041 DOI: 10.1007/s12288-023-01627-4] [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: 01/14/2022] [Accepted: 01/05/2023] [Indexed: 01/15/2023] Open
Abstract
Early T-cell precursor lymphoblastic leukemia (ETP-ALL) has a unique immunophenotype with very early T-cell differentiation. The current study summarises the distinct clinicopathological aspects of ETP-ALL and compares them with non-ETP-ALL. Twenty-nine ETP-ALL and 191 non-ETP-ALL cases were retrieved between 2018 and 2021. A P value was determined for each of the patient charaterisics (Table 1) to see for any significant relationship (P < 0.05) with ETP-ALL versus non-ETP-ALL. Kaplan-Meier log rank test was applied to look for any significant differences in OS for both the ALLs. ETP-ALL had an incidence of 12.6% out of total T-Acute lymphoblastic leukemia (T-ALL/LBL) in the past 3-years. Compared to non-ETP-ALL, ETP-ALL cases were associated with lower median age and male-to-female ratio. There was no statistically significant difference in the complete remission rate between both the subtypes. ETP-ALL was seen to be associated with high induction failure and relapse rate compared to non-ETP-ALL. To summarise, since the 2-year OS was poor compared to western research (for both ALLs), an intensive chemo-regimen should be implemented in the current situation. Some unusual markers were observed on flow-cytometry (ETP-ALL), which can be useful for MRD quantification, prognosis, and further trials for newer targeted therapies.
Collapse
Affiliation(s)
- Anurag Saha
- Department of Oncopathology, The Gujarat Cancer Research Institute, Asarwa, Ahmedabad, Gujarat 380016 India
| | - Beena Brahmbhatt
- Department of Oncopathology, The Gujarat Cancer Research Institute, Asarwa, Ahmedabad, Gujarat 380016 India
| | - Varnika Rai
- Department of Oncopathology, The Gujarat Cancer Research Institute, Asarwa, Ahmedabad, Gujarat 380016 India
| | - Sneha Kakoty
- Department of Oncopathology, The Gujarat Cancer Research Institute, Asarwa, Ahmedabad, Gujarat 380016 India
| | - Jyoti Sawhney
- Department of Oncopathology, The Gujarat Cancer Research Institute, Asarwa, Ahmedabad, Gujarat 380016 India
| |
Collapse
|
11
|
Liao C, Wang Y, Huang Y, Duan Y, Liang Y, Chen J, Jiang J, Shang K, Zhou C, Gu Y, Liu N, Zeng X, Gao X, Tang Y, Sun J. CD38-Specific CAR Integrated into CD38 Locus Driven by Different Promoters Causes Distinct Antitumor Activities of T and NK Cells. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2023; 10:e2207394. [PMID: 37485647 PMCID: PMC10520621 DOI: 10.1002/advs.202207394] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/13/2022] [Revised: 06/27/2023] [Indexed: 07/25/2023]
Abstract
The robust and stable expression of CD38 in T-cell acute lymphoblastic leukemia (T-ALL) blasts makes CD38 chimeric antigen receptor (CAR)-T/natural killer (NK) a potential therapy for T-ALL. However, CD38 expression in normal T/NK cells causes fratricide of CD38 CAR-T/NK cells. Here a "2-in-1" gene editing strategy is developed to generate fratricide-resistant locus-specific CAR-T/NK cells. CD38-specific CAR is integrated into the disrupted CD38 locus by CRISPR/Cas9, and CAR is placed under the control of either endogenous CD38 promoter (CD38KO/KI ) or exogenous EF1α promoter (CD38KO/KI EF1α). CD38 knockout reduces fratricide and allows the expansion of CAR-T cells. Meanwhile, CD38KO/KI EF1α results in higher CAR expression than CD38KO/KI in both CAR-T and CAR-NK cells. In a mouse T-ALL model, CD38KO/KI EF1α CAR-T cells eradicate tumors better than CD38KO/KI CAR-T cells. Surprisingly, CD38KO/KI CAR-NK cells show superior tumor control than CD38KO/KI EF1α CAR-NK cells. Further investigation reveals that endogenous regulatory elements in NK cells lead to higher expression of CD38 CAR than in T cells, and the expression levels of CAR affect the therapeutic outcome of CAR-T and CAR-NK cells differently. Therefore, these results support the efficacy of CD38 CAR-T/NK against T-ALL and demonstrate that the "2-in-1" strategy can resolve fratricide and enhance tumor eradication, paving the way for clinical translation.
Collapse
Affiliation(s)
- Chan Liao
- Department of Hematology‐oncologyChildren's HospitalZhejiang University School of MedicinePediatric Leukemia Diagnostic and Therapeutic Technology Research Center of Zhejiang Province National Clinical Research Center for Child HealthHangzhou310003China
| | - Yajie Wang
- Liangzhu LaboratoryZhejiang University Medical CenterHangzhou311121China
- Bone Marrow Transplantation Center of the First Affiliated Hospital and Department of Cell BiologyZhejiang University School of MedicineHangzhou310058China
- Institute of HematologyZhejiang University & Zhejiang Engineering Laboratory for Stem Cell and ImmunotherapyHangzhou310058China
| | - Yanjie Huang
- Key Laboratory of Structural Biology of Zhejiang ProvinceSchool of Life SciencesWestlake UniversityHangzhou310058China
- School of Basic Medical SciencesFudan UniversityShanghai200032China
| | - Yanting Duan
- Liangzhu LaboratoryZhejiang University Medical CenterHangzhou311121China
- Bone Marrow Transplantation Center of the First Affiliated Hospital and Department of Cell BiologyZhejiang University School of MedicineHangzhou310058China
- Institute of HematologyZhejiang University & Zhejiang Engineering Laboratory for Stem Cell and ImmunotherapyHangzhou310058China
| | - Yan Liang
- Liangzhu LaboratoryZhejiang University Medical CenterHangzhou311121China
| | - Jiangqing Chen
- Liangzhu LaboratoryZhejiang University Medical CenterHangzhou311121China
- Bone Marrow Transplantation Center of the First Affiliated Hospital and Department of Cell BiologyZhejiang University School of MedicineHangzhou310058China
- Institute of HematologyZhejiang University & Zhejiang Engineering Laboratory for Stem Cell and ImmunotherapyHangzhou310058China
| | - Jie Jiang
- Liangzhu LaboratoryZhejiang University Medical CenterHangzhou311121China
- Bone Marrow Transplantation Center of the First Affiliated Hospital and Department of Cell BiologyZhejiang University School of MedicineHangzhou310058China
- Institute of HematologyZhejiang University & Zhejiang Engineering Laboratory for Stem Cell and ImmunotherapyHangzhou310058China
| | - Kai Shang
- Liangzhu LaboratoryZhejiang University Medical CenterHangzhou311121China
- Bone Marrow Transplantation Center of the First Affiliated Hospital and Department of Cell BiologyZhejiang University School of MedicineHangzhou310058China
- Institute of HematologyZhejiang University & Zhejiang Engineering Laboratory for Stem Cell and ImmunotherapyHangzhou310058China
| | - Chun Zhou
- School of Public Health and Sir Run Run Shaw HospitalZhejiang University School of MedicineHangzhou310058China
| | - Ying Gu
- Institute of Genetics, Zhejiang University and Department of GeneticsZhejiang University school of medicineHangzhou310058China
| | - Nan Liu
- Liangzhu LaboratoryZhejiang University Medical CenterHangzhou311121China
| | - Xun Zeng
- State Key Laboratory for Diagnosis and Treatment of Infectious DiseasesFirst Affiliated HospitalZhejiang University School of MedicineHangzhou310058China
| | - Xiaofei Gao
- Key Laboratory of Structural Biology of Zhejiang ProvinceSchool of Life SciencesWestlake UniversityHangzhou310058China
| | - Yongmin Tang
- Department of Hematology‐oncologyChildren's HospitalZhejiang University School of MedicinePediatric Leukemia Diagnostic and Therapeutic Technology Research Center of Zhejiang Province National Clinical Research Center for Child HealthHangzhou310003China
| | - Jie Sun
- Liangzhu LaboratoryZhejiang University Medical CenterHangzhou311121China
- Bone Marrow Transplantation Center of the First Affiliated Hospital and Department of Cell BiologyZhejiang University School of MedicineHangzhou310058China
- Institute of HematologyZhejiang University & Zhejiang Engineering Laboratory for Stem Cell and ImmunotherapyHangzhou310058China
| |
Collapse
|
12
|
Tapia-Galisteo A, Álvarez-Vallina L, Sanz L. Bi- and trispecific immune cell engagers for immunotherapy of hematological malignancies. J Hematol Oncol 2023; 16:83. [PMID: 37501154 PMCID: PMC10373336 DOI: 10.1186/s13045-023-01482-w] [Citation(s) in RCA: 44] [Impact Index Per Article: 22.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2023] [Accepted: 07/19/2023] [Indexed: 07/29/2023] Open
Abstract
Immune cell engagers are engineered antibodies with at least one arm binding a tumor-associated antigen and at least another one directed against an activating receptor in immune effector cells: CD3 for recruitment of T cells and CD16a for NK cells. The first T cell engager (the anti-CD19 blinatumomab) was approved by the FDA in 2014, but no other one hit the market until 2022. Now the field is gaining momentum, with three approvals in 2022 and 2023 (as of May): the anti-CD20 × anti-CD3 mosunetuzumab and epcoritamab and the anti-B cell maturation antigen (BCMA) × anti-CD3 teclistamab, and another three molecules in regulatory review. T cell engagers will likely revolutionize the treatment of hematological malignancies in the short term, as they are considerably more potent than conventional monoclonal antibodies recognizing the same tumor antigens. The field is thriving, with a plethora of different formats and targets, and around 100 bispecific T cell engagers more are already in clinical trials. Bispecific NK cell engagers are also in early-stage clinical studies and may offer similar efficacy with milder side effects. Trispecific antibodies (engaging either T cell or NK cell receptors) raise the game even further with a third binding moiety, which allows either the targeting of an additional tumor-associated antigen to increase specificity and avoid immune escape or the targeting of additional costimulatory receptors on the immune cell to improve its effector functions. Altogether, these engineered molecules may change the paradigm of treatment for relapsed or refractory hematological malignancies.
Collapse
Affiliation(s)
- Antonio Tapia-Galisteo
- Immuno-Oncology and Immunotherapy Group, Biomedical Research Institute Hospital Universitario, 12 de Octubre, Madrid, Spain
- Cancer Immunotherapy Unit (UNICA), Department of Immunology, Hospital Universitario, 12 de Octubre, Madrid, Spain
- H12O-CNIO Cancer Immunotherapy Clinical Research Unit, Centro Nacional de Investigaciones Oncológicas (CNIO), Madrid, Spain
| | - Luis Álvarez-Vallina
- Immuno-Oncology and Immunotherapy Group, Biomedical Research Institute Hospital Universitario, 12 de Octubre, Madrid, Spain.
- Cancer Immunotherapy Unit (UNICA), Department of Immunology, Hospital Universitario, 12 de Octubre, Madrid, Spain.
- H12O-CNIO Cancer Immunotherapy Clinical Research Unit, Centro Nacional de Investigaciones Oncológicas (CNIO), Madrid, Spain.
| | - Laura Sanz
- Molecular Immunology Unit, Biomedical Research Institute Hospital Universitario Puerta de Hierro Majadahonda, Madrid, Spain.
| |
Collapse
|
13
|
Moles E, Howard CB, Huda P, Karsa M, McCalmont H, Kimpton K, Duly A, Chen Y, Huang Y, Tursky ML, Ma D, Bustamante S, Pickford R, Connerty P, Omari S, Jolly CJ, Joshi S, Shen S, Pimanda JE, Dolnikov A, Cheung LC, Kotecha RS, Norris MD, Haber M, de Bock CE, Somers K, Lock RB, Thurecht KJ, Kavallaris M. Delivery of PEGylated liposomal doxorubicin by bispecific antibodies improves treatment in models of high-risk childhood leukemia. Sci Transl Med 2023; 15:eabm1262. [PMID: 37196067 DOI: 10.1126/scitranslmed.abm1262] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2021] [Accepted: 04/13/2023] [Indexed: 05/19/2023]
Abstract
High-risk childhood leukemia has a poor prognosis because of treatment failure and toxic side effects of therapy. Drug encapsulation into liposomal nanocarriers has shown clinical success at improving biodistribution and tolerability of chemotherapy. However, enhancements in drug efficacy have been limited because of a lack of selectivity of the liposomal formulations for the cancer cells. Here, we report on the generation of bispecific antibodies (BsAbs) with dual binding to a leukemic cell receptor, such as CD19, CD20, CD22, or CD38, and methoxy polyethylene glycol (PEG) for the targeted delivery of PEGylated liposomal drugs to leukemia cells. This liposome targeting system follows a "mix-and-match" principle where BsAbs were selected on the specific receptors expressed on leukemia cells. BsAbs improved the targeting and cytotoxic activity of a clinically approved and low-toxic PEGylated liposomal formulation of doxorubicin (Caelyx) toward leukemia cell lines and patient-derived samples that are immunophenotypically heterogeneous and representative of high-risk subtypes of childhood leukemia. BsAb-assisted improvements in leukemia cell targeting and cytotoxic potency of Caelyx correlated with receptor expression and were minimally detrimental in vitro and in vivo toward expansion and functionality of normal peripheral blood mononuclear cells and hematopoietic progenitors. Targeted delivery of Caelyx using BsAbs further enhanced leukemia suppression while reducing drug accumulation in the heart and kidneys and extended overall survival in patient-derived xenograft models of high-risk childhood leukemia. Our methodology using BsAbs therefore represents an attractive targeting platform to potentiate the therapeutic efficacy and safety of liposomal drugs for improved treatment of high-risk leukemia.
Collapse
Affiliation(s)
- Ernest Moles
- Children's Cancer Institute, Lowy Cancer Research Centre, UNSW Sydney, Sydney 2052, Australia
- Australian Centre for Nanomedicine, Faculty of Engineering, UNSW Sydney, Sydney 2052, Australia
- School of Clinical Medicine, Medicine and Health, UNSW Sydney, Sydney 2052, Australia
| | - Christopher B Howard
- Australian Institute for Bioengineering and Nanotechnology, University of Queensland, St Lucia 4072, Australia
| | - Pie Huda
- Australian Institute for Bioengineering and Nanotechnology, University of Queensland, St Lucia 4072, Australia
| | - Mawar Karsa
- Children's Cancer Institute, Lowy Cancer Research Centre, UNSW Sydney, Sydney 2052, Australia
- School of Clinical Medicine, Medicine and Health, UNSW Sydney, Sydney 2052, Australia
| | - Hannah McCalmont
- Children's Cancer Institute, Lowy Cancer Research Centre, UNSW Sydney, Sydney 2052, Australia
- School of Clinical Medicine, Medicine and Health, UNSW Sydney, Sydney 2052, Australia
| | - Kathleen Kimpton
- Children's Cancer Institute, Lowy Cancer Research Centre, UNSW Sydney, Sydney 2052, Australia
- School of Clinical Medicine, Medicine and Health, UNSW Sydney, Sydney 2052, Australia
| | - Alastair Duly
- Children's Cancer Institute, Lowy Cancer Research Centre, UNSW Sydney, Sydney 2052, Australia
- School of Clinical Medicine, Medicine and Health, UNSW Sydney, Sydney 2052, Australia
| | - Yongjuan Chen
- Children's Cancer Institute, Lowy Cancer Research Centre, UNSW Sydney, Sydney 2052, Australia
- School of Clinical Medicine, Medicine and Health, UNSW Sydney, Sydney 2052, Australia
| | - Yizhou Huang
- Children's Cancer Institute, Lowy Cancer Research Centre, UNSW Sydney, Sydney 2052, Australia
- School of Clinical Medicine, Medicine and Health, UNSW Sydney, Sydney 2052, Australia
| | - Melinda L Tursky
- Department of Haematology and Bone Marrow Transplant, St Vincent's Hospital Sydney, Sydney 2010, Australia
- St Vincent's Centre for Applied Medical Research (AMR), Sydney 2010, Australia
- St Vincent Clinical School, Faculty of Medicine and Health, UNSW Sydney, Sydney 2052, Australia
| | - David Ma
- Department of Haematology and Bone Marrow Transplant, St Vincent's Hospital Sydney, Sydney 2010, Australia
- St Vincent's Centre for Applied Medical Research (AMR), Sydney 2010, Australia
- St Vincent Clinical School, Faculty of Medicine and Health, UNSW Sydney, Sydney 2052, Australia
| | - Sonia Bustamante
- Bioanalytical Mass Spectrometry Facility, Mark Wainwright Analytical Centre, UNSW Sydney, Sydney 2052, Australia
| | - Russell Pickford
- Bioanalytical Mass Spectrometry Facility, Mark Wainwright Analytical Centre, UNSW Sydney, Sydney 2052, Australia
| | - Patrick Connerty
- Children's Cancer Institute, Lowy Cancer Research Centre, UNSW Sydney, Sydney 2052, Australia
- School of Clinical Medicine, Medicine and Health, UNSW Sydney, Sydney 2052, Australia
| | - Sofia Omari
- Children's Cancer Institute, Lowy Cancer Research Centre, UNSW Sydney, Sydney 2052, Australia
- School of Clinical Medicine, Medicine and Health, UNSW Sydney, Sydney 2052, Australia
| | - Christopher J Jolly
- School of Biomedical Sciences, Lowy Cancer Research Centre, UNSW Sydney, Sydney 2052, Australia
| | - Swapna Joshi
- School of Biomedical Sciences, Lowy Cancer Research Centre, UNSW Sydney, Sydney 2052, Australia
| | - Sylvie Shen
- School of Biomedical Sciences, Lowy Cancer Research Centre, UNSW Sydney, Sydney 2052, Australia
| | - John E Pimanda
- School of Clinical Medicine, Medicine and Health, UNSW Sydney, Sydney 2052, Australia
- School of Biomedical Sciences, Lowy Cancer Research Centre, UNSW Sydney, Sydney 2052, Australia
- Department of Haematology, Prince of Wales Hospital, Sydney 2031, Australia
| | - Alla Dolnikov
- Children's Cancer Institute, Lowy Cancer Research Centre, UNSW Sydney, Sydney 2052, Australia
- School of Clinical Medicine, Medicine and Health, UNSW Sydney, Sydney 2052, Australia
| | - Laurence C Cheung
- Leukaemia Translational Research Laboratory, Telethon Kids Cancer Centre, Telethon Kids Institute, Perth, Western Australia 6009, Australia
- Curtin Medical School, Curtin University, Perth, Western Australia 6102, Australia
| | - Rishi S Kotecha
- Leukaemia Translational Research Laboratory, Telethon Kids Cancer Centre, Telethon Kids Institute, Perth, Western Australia 6009, Australia
- Curtin Medical School, Curtin University, Perth, Western Australia 6102, Australia
- Department of Clinical Haematology, Oncology, Blood and Marrow Transplantation, Perth Children's Hospital, Perth, Western Australia 6009, Australia
- School of Medicine, University of Western Australia, Perth, Western Australia 6009, Australia
| | - Murray D Norris
- Children's Cancer Institute, Lowy Cancer Research Centre, UNSW Sydney, Sydney 2052, Australia
- School of Clinical Medicine, Medicine and Health, UNSW Sydney, Sydney 2052, Australia
- University of New South Wales Centre for Childhood Cancer Research, UNSW Sydney, Sydney 2052, Australia
| | - Michelle Haber
- Children's Cancer Institute, Lowy Cancer Research Centre, UNSW Sydney, Sydney 2052, Australia
- School of Clinical Medicine, Medicine and Health, UNSW Sydney, Sydney 2052, Australia
| | - Charles E de Bock
- Children's Cancer Institute, Lowy Cancer Research Centre, UNSW Sydney, Sydney 2052, Australia
- School of Clinical Medicine, Medicine and Health, UNSW Sydney, Sydney 2052, Australia
| | - Klaartje Somers
- Children's Cancer Institute, Lowy Cancer Research Centre, UNSW Sydney, Sydney 2052, Australia
- School of Clinical Medicine, Medicine and Health, UNSW Sydney, Sydney 2052, Australia
| | - Richard B Lock
- Children's Cancer Institute, Lowy Cancer Research Centre, UNSW Sydney, Sydney 2052, Australia
- School of Clinical Medicine, Medicine and Health, UNSW Sydney, Sydney 2052, Australia
| | - Kristofer J Thurecht
- Australian Institute for Bioengineering and Nanotechnology, University of Queensland, St Lucia 4072, Australia
- Centre for Advanced Imaging, ARC Training Centre for Innovation in Biomedical Imaging Technologies, University of Queensland, St Lucia 4072, Australia
| | - Maria Kavallaris
- Children's Cancer Institute, Lowy Cancer Research Centre, UNSW Sydney, Sydney 2052, Australia
- Australian Centre for Nanomedicine, Faculty of Engineering, UNSW Sydney, Sydney 2052, Australia
- School of Clinical Medicine, Medicine and Health, UNSW Sydney, Sydney 2052, Australia
| |
Collapse
|
14
|
Chu Y, Gardenswartz A, Diorio C, Marks LJ, Lowe E, Teachey DT, Cairo MS. Cellular and humoral immunotherapy in children, adolescents and young adults with non-Hodgkin lymphoma. Best Pract Res Clin Haematol 2023; 36:101442. [PMID: 36907635 DOI: 10.1016/j.beha.2023.101442] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2023] [Accepted: 01/31/2023] [Indexed: 02/05/2023]
Abstract
The prognosis is dismal (2-year overall survival less than 25%) for childhood, adolescent, and young adult (CAYA) with relapsed and/or refractory (R/R) non-Hodgkin lymphoma (NHL). Novel targeted therapies are desperately needed for this poor-risk population. CD19, CD20, CD22, CD79a, CD38, CD30, LMP1 and LMP2 are attractive targets for immunotherapy in CAYA patients with R/R NHL. Novel anti-CD20 monoclonal antibodies, anti-CD38 monoclonal antibody, antibody drug conjugates and T and natural killer (NK)-cell bispecific and trispecific engagers are being investigated in the R/R setting and are changing the landscape of NHL therapy. A variety of cellular immunotherapies such as viral activated cytotoxic T-lymphocyte, chimeric antigen receptor (CAR) T-cells, NK and CAR NK-cells have been investigated and provide alternative options for CAYA patients with R/R NHL. Here, we provide an update and clinical practice guidance of utilizing these cellular and humoral immunotherapies in CAYA patients with R/R NHL.
Collapse
Affiliation(s)
- Yaya Chu
- Department of Pediatrics, New York Medical College, Valhalla, NY, USA
| | | | - Caroline Diorio
- The Children's Hospital of Philadelphia, Philadelphia, PA, USA
| | - Lianna J Marks
- Division of Pediatric Hematology and Oncology, Stanford University School of Medicine, Palo Alto, CA, USA
| | - Eric Lowe
- Division of Pediatric Hematology-Oncology, Children's Hospital of the Kings Daughter, Norfolk, VA, USA
| | - David T Teachey
- The Children's Hospital of Philadelphia, Philadelphia, PA, USA; University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
| | - Mitchell S Cairo
- Department of Pediatrics, New York Medical College, Valhalla, NY, USA; Department of Epidemiology and Community Health, New York Medical College, Valhalla, NY, USA; Department of Medicine, New York Medical College, Valhalla, NY, USA; Department of Pathology, Microbiology and Immunology, New York Medical College, Valhalla, NY, USA; Department of Cell Biology, New York Medical College, Valhalla, NY, USA; Department of Anatomy, New York Medical College, Valhalla, NY, USA.
| |
Collapse
|
15
|
Mills CM, Benton TZ, Piña I, Francis MJ, Reyes L, Dolloff NG, Peterson YK, Woster PM. Stimulation of natural killer cells with small molecule inhibitors of CD38 for the treatment of neuroblastoma. Chem Sci 2023; 14:2168-2182. [PMID: 36845935 PMCID: PMC9945084 DOI: 10.1039/d2sc05749b] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2022] [Accepted: 01/28/2023] [Indexed: 02/02/2023] Open
Abstract
High-risk neuroblastoma (NB) accounts for 15% of all pediatric cancer deaths. Refractory disease for high-risk NB patients is attributed to chemotherapy resistance and immunotherapy failure. The poor prognosis for high-risk NB patients demonstrates an unmet medical need for the development of new, more efficacious therapeutics. CD38 is an immunomodulating protein that is expressed constitutively on natural killer (NK) cells and other immune cells in the tumor microenvironment (TME). Furthermore, CD38 over expression is implicated in propagating an immunosuppressive milieu within the TME. Through virtual and physical screening, we have identified drug-like small molecule inhibitors of CD38 with low micromolar IC50 values. We have begun to explore structure activity relationships for CD38 inhibition through derivatization of our most effective hit molecule to develop a new compound with lead-like physicochemical properties and improved potency. We have demonstrated that our derivatized inhibitor, compound 2, elicits immunomodulatory effects in NK cells by increasing cell viability by 190 ± 36% in multiple donors and by significantly increasing interferon gamma. Additionally, we have illustrated that NK cells exhibited enhanced cytotoxicity toward NB cells (14% reduction of NB cells over 90 minutes) when given a combination treatment of our inhibitor and the immunocytokine ch14.18-IL2. Herein we describe the synthesis and biological evaluation of small molecule CD38 inhibitors and demonstrate their potential utility as a novel approach to NB immunotherapy. These compounds represent the first examples of small molecules that stimulate immune function for the treatment of cancer.
Collapse
Affiliation(s)
- Catherine M Mills
- Department of Drug Discovery and Biomedical Sciences, Medical University of South Carolina 70 President St Charleston SC 29425 USA
| | - Thomas Z Benton
- Department of Drug Discovery and Biomedical Sciences, Medical University of South Carolina 70 President St Charleston SC 29425 USA
| | - Ivett Piña
- Department of Drug Discovery and Biomedical Sciences, Medical University of South Carolina 70 President St Charleston SC 29425 USA
| | - Megan J Francis
- Department of Drug Discovery and Biomedical Sciences, Medical University of South Carolina 70 President St Charleston SC 29425 USA
| | - Leticia Reyes
- Department of Cell and Molecular Pharmacology and Experimental Therapeutics, Medical University of South Carolina 70 President St Charleston SC 29425 USA
| | - Nathan G Dolloff
- Department of Cell and Molecular Pharmacology and Experimental Therapeutics, Medical University of South Carolina 70 President St Charleston SC 29425 USA
| | - Yuri K Peterson
- Department of Drug Discovery and Biomedical Sciences, Medical University of South Carolina 70 President St Charleston SC 29425 USA
| | - Patrick M Woster
- Department of Drug Discovery and Biomedical Sciences, Medical University of South Carolina 70 President St Charleston SC 29425 USA
| |
Collapse
|
16
|
Varadarajan I, Pierce E, Scheuing L, Morris A, El Chaer F, Keng M. Post-Hematopoietic Cell Transplantation Relapsed Acute Lymphoblastic Leukemia: Current Challenges and Future Directions. Onco Targets Ther 2023; 16:1-16. [PMID: 36685611 PMCID: PMC9849790 DOI: 10.2147/ott.s274551] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2022] [Accepted: 12/14/2022] [Indexed: 01/15/2023] Open
Abstract
Allogeneic hematopoietic cell transplantation (allo-HCT) represents an important and potentially curative treatment option for adult patients with acute lymphoblastic leukemia. Relapse continues to remain the most important factor influencing overall survival post allo-HCT. We discuss early identification, clinical manifestations, and management of relapsed disease. Routine evaluation of measurable residual disease (MRD) and change in donor chimerism play a crucial role in early detection. Pivotal clinical trials have led to FDA approval of multiple novel agents like blinatumomab and inotuzumab. Combining targeted therapy with cellular immunotherapy serves as the backbone for prolonging overall survival in these patients. Donor lymphocyte infusions have traditionally been used in relapsed disease with suboptimal outcomes. This review provides insight into use of cellular therapy in MRD positivity and decreasing donor chimerism. It also discusses various modalities of combining cellular therapy with novel agents and discussing the impact of chimeric antigen receptor T-cell therapy in the setting of post allo-HCT relapse both as consolidative therapy and as a bridge to second transplant.
Collapse
Affiliation(s)
- Indumathy Varadarajan
- Department of Medicine, Division of Hematology and Oncology, University of Virginia, Charlottesville, VA, USA
| | - Eric Pierce
- Department of Medicine, Division of Hematology and Oncology, University of Virginia, Charlottesville, VA, USA
| | - Lisa Scheuing
- Department of Medicine, Division of Hematology and Oncology, University of Virginia, Charlottesville, VA, USA
| | - Amy Morris
- Department of Pharmacy Services, University of Virginia, Charlottesville, VA, USA
| | - Firas El Chaer
- Department of Medicine, Division of Hematology and Oncology, University of Virginia, Charlottesville, VA, USA
| | - Michael Keng
- Department of Medicine, Division of Hematology and Oncology, University of Virginia, Charlottesville, VA, USA,Correspondence: Michael Keng, Division of Hematology & Oncology, University of Virginia Comprehensive Cancer Center, West Complex Room 6009, 1300 Jefferson Park Ave, PO Box 800716, Charlottesville, VA, 22908, USA, Tel +1 434 924 4257, Fax +1 434- 243 6068, Email
| |
Collapse
|
17
|
Caracciolo D, Mancuso A, Polerà N, Froio C, D'Aquino G, Riillo C, Tagliaferri P, Tassone P. The emerging scenario of immunotherapy for T-cell Acute Lymphoblastic Leukemia: advances, challenges and future perspectives. Exp Hematol Oncol 2023; 12:5. [PMID: 36624522 PMCID: PMC9828428 DOI: 10.1186/s40164-022-00368-w] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2022] [Accepted: 12/30/2022] [Indexed: 01/11/2023] Open
Abstract
T-cell acute lymphoblastic leukemia (T-ALL) is a challenging pediatric and adult haematologic disease still associated with an unsatisfactory cure rate. Unlike B-ALL, the availability of novel therapeutic options to definitively improve the life expectancy for relapsed/resistant patients is poor. Indeed, the shared expression of surface targets among normal and neoplastic T-cells still limits the efficacy and may induce fratricide effects, hampering the use of innovative immunotherapeutic strategies. However, novel monoclonal antibodies, bispecific T-cell engagers (BTCEs), and chimeric antigen receptors (CAR) T-cells recently showed encouraging results and some of them are in an advanced stage of pre-clinical development or are currently under investigation in clinical trials. Here, we review this exciting scenario focusing on most relevant advances, challenges, and perspectives of the emerging landscape of immunotherapy of T-cell malignancies.
Collapse
Affiliation(s)
- Daniele Caracciolo
- Department of Experimental and Clinical Medicine, Magna Græcia University, Catanzaro, Italy
| | - Antonia Mancuso
- Department of Experimental and Clinical Medicine, Magna Græcia University, Catanzaro, Italy
| | - Nicoletta Polerà
- Department of Experimental and Clinical Medicine, Magna Græcia University, Catanzaro, Italy
| | - Caterina Froio
- Department of Experimental and Clinical Medicine, Magna Græcia University, Catanzaro, Italy
| | - Giuseppe D'Aquino
- Department of Experimental and Clinical Medicine, Magna Græcia University, Catanzaro, Italy
| | - Caterina Riillo
- Department of Experimental and Clinical Medicine, Magna Græcia University, Catanzaro, Italy
| | | | - Pierfrancesco Tassone
- Department of Experimental and Clinical Medicine, Magna Græcia University, Catanzaro, Italy.
- Sbarro Institute for Cancer Research and Molecular Medicine, Center for Biotechnology, College of Science and Technology, Temple University, Philadelphia, PA, USA.
| |
Collapse
|
18
|
Thomas X. T-cell acute lymphoblastic leukemia: promising experimental drugs in clinical development. Expert Opin Investig Drugs 2023; 32:37-52. [PMID: 36541671 DOI: 10.1080/13543784.2023.2161361] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
INTRODUCTION Despite advances in treatment approaches in acute lymphoblastic leukemia (ALL), the prognosis of adults with newly diagnosed T-ALL remains poor, as well as that of adults and children with relapsed disease. Novel targeted therapies are therefore needed. AREAS COVERED This review summarizes promising emerging strategies for the treatment of T-ALL. EXPERT OPINION The recent molecular characterization of T-ALL has led to the identification of new therapeutic targets. Small-molecules inhibitors and other targeted therapies have therefore been recently developed and are currently under clinical investigations. Similarly, first studies involving monoclonal antibodies and chimeric antigen receptor (CAR) T cells have shown encouraging results. Improvement of outcome with these novel approaches, eventually combined with current standard chemotherapy, is therefore expected in a near future in T-ALL.
Collapse
Affiliation(s)
- Xavier Thomas
- Hospices Civils de Lyon, Department of Clinical Hematology, Centre Hospitalier Lyon-Sud, Pierre Bénite, France
| |
Collapse
|
19
|
Brum da Silva Nunes V, Kehl Dias C, Nathali Scholl J, Nedel Sant'Ana A, de Fraga Dias A, Granero Farias M, Alegretti AP, Sosnoski M, Esteves Daudt L, Bohns Michalowski M, Oliveira Battastini AM, Paz AA, Figueiró F. Lymphocytes from B-acute lymphoblastic leukemia patients present differential regulation of the adenosinergic axis depending on risk stratification. Discov Oncol 2022; 13:143. [PMID: 36581667 PMCID: PMC9800668 DOI: 10.1007/s12672-022-00602-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/18/2022] [Accepted: 12/08/2022] [Indexed: 12/31/2022] Open
Abstract
PURPOSE Although risk-stratified chemotherapy regimens improve B-cell acute lymphoblastic leukemia (B-ALL) clinical outcome, relapse occurs in a significant number of cases. The identification of new therapeutic targets as well as prognostic and diagnostic biomarkers can improve B-ALL patients' clinical outcomes. Purinergic signaling is an important pathway in cancer progression, however the expression of ectonucleotidases and their impact on immune cells in B-ALL lacks exploration. We aimed to analyze the expression of ectonucleotidases in B-ALL patients' lymphocyte subpopulations. METHODS Peripheral blood samples from 15 patients diagnosed with B-ALL were analyzed. Flow cytometry was used to analyze cellularity, expression level of CD38, CD39, and CD73, and frequency of [Formula: see text], and [Formula: see text] in lymphocyte subpopulations. Plasma was used for cytokines (by CBA kit) and adenine nucleosides/nucleotides detection (by HPLC). RESULTS Comparing B-ALL patients to health donors, we observed an increase of CD4 + and CD8 + T-cells. In addition, a decrease in CD38 expression in B and Treg subpopulations and an increase in CD39+ CD73+ frequency in Breg and CD8+ T-cells. Analyzing cytokines and adenine nucleosides/nucleotides, we found a decrease in TNF, IL-1β, and ADO concentrations, together with an increase in AMP in B-ALL patients' plasma. CONCLUSION As immunomodulators, the expression of ectonucleotidases might be associated with activation states, as well as the abundance of different cellular subsets. We observed a pro-tumor immunity expression profile in B-ALL patients at diagnosis, being associated with cell exhaustion and immune evasion in B-ALL.
Collapse
Affiliation(s)
- Vitória Brum da Silva Nunes
- Laboratório de Imunobioquímica do Câncer, Departamento de Bioquímica, Instituto de Ciências Básicas da Saúde, UFRGS, Porto Alegre, RS, CEP 90035-003, Brazil
- Programa de Pós-Graduação em Ciências Biológicas: Bioquímica, Instituto de Ciências Básicas da Saúde, UFRGS, Porto Alegre, RS, CEP 90035-003, Brazil
| | - Camila Kehl Dias
- Laboratório de Imunobioquímica do Câncer, Departamento de Bioquímica, Instituto de Ciências Básicas da Saúde, UFRGS, Porto Alegre, RS, CEP 90035-003, Brazil
- Programa de Pós-Graduação em Ciências Biológicas: Bioquímica, Instituto de Ciências Básicas da Saúde, UFRGS, Porto Alegre, RS, CEP 90035-003, Brazil
| | - Juliete Nathali Scholl
- Laboratório de Imunobioquímica do Câncer, Departamento de Bioquímica, Instituto de Ciências Básicas da Saúde, UFRGS, Porto Alegre, RS, CEP 90035-003, Brazil
- Programa de Pós-Graduação em Ciências Biológicas: Bioquímica, Instituto de Ciências Básicas da Saúde, UFRGS, Porto Alegre, RS, CEP 90035-003, Brazil
| | - Alexia Nedel Sant'Ana
- Laboratório de Imunobioquímica do Câncer, Departamento de Bioquímica, Instituto de Ciências Básicas da Saúde, UFRGS, Porto Alegre, RS, CEP 90035-003, Brazil
- Programa de Pós-Graduação em Ciências Biológicas: Bioquímica, Instituto de Ciências Básicas da Saúde, UFRGS, Porto Alegre, RS, CEP 90035-003, Brazil
| | - Amanda de Fraga Dias
- Laboratório de Imunobioquímica do Câncer, Departamento de Bioquímica, Instituto de Ciências Básicas da Saúde, UFRGS, Porto Alegre, RS, CEP 90035-003, Brazil
- Programa de Pós-Graduação em Ciências Biológicas: Bioquímica, Instituto de Ciências Básicas da Saúde, UFRGS, Porto Alegre, RS, CEP 90035-003, Brazil
| | | | - Ana Paula Alegretti
- Hospital de Clínicas de Porto Alegre/HCPA, Porto Alegre, RS, CEP 90035-903, Brazil
| | - Monalisa Sosnoski
- Hospital de Clínicas de Porto Alegre/HCPA, Porto Alegre, RS, CEP 90035-903, Brazil
| | - Liane Esteves Daudt
- Hospital de Clínicas de Porto Alegre/HCPA, Porto Alegre, RS, CEP 90035-903, Brazil
| | - Mariana Bohns Michalowski
- Hospital de Clínicas de Porto Alegre/HCPA, Porto Alegre, RS, CEP 90035-903, Brazil
- Programa de Pós-Graduação em Saúde da Criança e do Adolescente, Faculdade de Medicina, UFRGS, Porto Alegre, RS, 90035-003, Brazil
| | - Ana Maria Oliveira Battastini
- Laboratório de Imunobioquímica do Câncer, Departamento de Bioquímica, Instituto de Ciências Básicas da Saúde, UFRGS, Porto Alegre, RS, CEP 90035-003, Brazil
- Programa de Pós-Graduação em Ciências Biológicas: Bioquímica, Instituto de Ciências Básicas da Saúde, UFRGS, Porto Alegre, RS, CEP 90035-003, Brazil
| | | | - Fabrício Figueiró
- Laboratório de Imunobioquímica do Câncer, Departamento de Bioquímica, Instituto de Ciências Básicas da Saúde, UFRGS, Porto Alegre, RS, CEP 90035-003, Brazil.
- Programa de Pós-Graduação em Ciências Biológicas: Bioquímica, Instituto de Ciências Básicas da Saúde, UFRGS, Porto Alegre, RS, CEP 90035-003, Brazil.
| |
Collapse
|
20
|
Zhang Z, Yang K, Zhang H. Targeting Leukemia-Initiating Cells and Leukemic Niches: The Next Therapy Station for T-Cell Acute Lymphoblastic Leukemia? Cancers (Basel) 2022; 14:cancers14225655. [PMID: 36428753 PMCID: PMC9688677 DOI: 10.3390/cancers14225655] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2022] [Revised: 11/09/2022] [Accepted: 11/15/2022] [Indexed: 11/19/2022] Open
Abstract
T-cell acute lymphoblastic leukemia (T-ALL) is an aggressive subtype of hematological malignancy characterized by its high heterogeneity and potentially life-threatening clinical features. Despite the advances in risk stratification and therapeutic management of T-ALL, patients often suffer from treatment failure and chemotherapy-induced toxicity, calling for greater efforts to improve therapeutic efficacy and safety in the treatment of T-ALL. During the past decades, increasing evidence has shown the indispensable effects of leukemia-initiating cells (LICs) and leukemic niches on T-ALL initiation and progression. These milestones greatly facilitate precision medicine by interfering with the pathways that are associated with LICs and leukemic niches or by targeting themselves directly. Most of these novel agents, either alone or in combination with conventional chemotherapy, have shown promising preclinical results, facilitating them to be further evaluated under clinical trials. In this review, we summarize the latest discoveries in LICs and leukemic niches in terms of T-ALL, with a particular highlight on the current precision medicine. The challenges and future prospects are also discussed.
Collapse
Affiliation(s)
- Ziting Zhang
- Institute of Medical Biology, Chinese Academy of Medical Sciences and Peking Union Medical College, Kunming 650118, China
| | - Kun Yang
- Institute of Medical Biology, Chinese Academy of Medical Sciences and Peking Union Medical College, Kunming 650118, China
- School of Life Sciences, Yunnan University, Kunming 650500, China
| | - Han Zhang
- Institute of Medical Biology, Chinese Academy of Medical Sciences and Peking Union Medical College, Kunming 650118, China
- Correspondence: ; Tel.: +86-158-7796-3252
| |
Collapse
|
21
|
Sriram H, Kunjachan F, Khanka T, Gawai S, Ghogale S, Deshpande N, Girase K, Patil J, Chatterjee G, Rajpal S, Patkar NV, Bagal B, Jain H, Sengar M, Hasan SK, Khattry N, Subramanian PG, Gujral S, Tembhare PR. Expression levels and patterns of B-cell maturation antigen in newly diagnosed and relapsed multiple myeloma patients from Indian subcontinent. CYTOMETRY. PART B, CLINICAL CYTOMETRY 2022; 102:462-470. [PMID: 36346307 DOI: 10.1002/cyto.b.22099] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/19/2022] [Revised: 09/16/2022] [Accepted: 10/18/2022] [Indexed: 11/09/2022]
Abstract
BACKGROUND Many novel therapies are being evaluated for the treatment of Multiple myeloma (MM). The cell-surface protein B-cell maturation antigen (BCMA, CD269) has recently emerged as a promising target for CAR-T cell and monoclonal-antibody therapies in MM. However, the knowledge of the BCMA expression-pattern in myeloma patients from the Indian subcontinent is still not available. We present an in-depth study of BCMA expression-pattern on abnormal plasma cells (aPC) in Indian MM patients. METHODS We studied BM samples from 217 MM patients (211-new and 6-relapsed) with a median age of 56 years (range, 30-78 years & M:F-2.29) and 20 control samples. Expression levels/patterns of CD269 (clone-19f2) were evaluated in aPCs from MM patients and in normal PCs (nPC) from uninvolved staging bone marrow samples (controls) using multicolor flow cytometry (MFC). Expression-level of CD269 was determined as a ratio of mean fluorescent intensity (MFI-R) of CD269 in PCs to that of non-B-lymphocytes and expression-pattern (homogenous/heterogeneous) as coefficient-of-variation of immunofluorescence (CVIF). RESULTS Median (range) percentage of CD269-positive abnormal-PCs in total PCs was 71.6% (0.49-99.29%). The MFI-R (median, range) of CD269 was significantly higher in aPCs (4.13, 1.12-26.88) than nPCs (3.33, 1.23-12.87), p < .0001. Median (range) MFI of CD269 at diagnosis and relapse were 2.39 (0.77-9.57) and 2.66 (2.15-3.23) respectively. CD269 levels were similar at diagnosis and relapse, p = .5529. CONCLUSIONS We demonstrated that BCMA/CD269 is highly expressed in aPCs from a majority of MM patients, both at diagnosis and relapse. Thus, BCMA is a valuable target for therapy for Indian MM patients.
Collapse
Affiliation(s)
- Harshini Sriram
- Hematopathology Laboratory, ACTREC, Tata Memorial Centre, Homi Bhabha National Institute, Mumbai, India
| | - Florence Kunjachan
- Hematopathology Laboratory, ACTREC, Tata Memorial Centre, Homi Bhabha National Institute, Mumbai, India
| | - Twinkle Khanka
- Hematopathology Laboratory, ACTREC, Tata Memorial Centre, Homi Bhabha National Institute, Mumbai, India
| | - Sangamitra Gawai
- Hematopathology Laboratory, ACTREC, Tata Memorial Centre, Homi Bhabha National Institute, Mumbai, India
| | - Sitaram Ghogale
- Hematopathology Laboratory, ACTREC, Tata Memorial Centre, Homi Bhabha National Institute, Mumbai, India
| | - Nilesh Deshpande
- Hematopathology Laboratory, ACTREC, Tata Memorial Centre, Homi Bhabha National Institute, Mumbai, India
| | - Karishma Girase
- Hematopathology Laboratory, ACTREC, Tata Memorial Centre, Homi Bhabha National Institute, Mumbai, India
| | - Jagruti Patil
- Hematopathology Laboratory, ACTREC, Tata Memorial Centre, Homi Bhabha National Institute, Mumbai, India
| | - Gaurav Chatterjee
- Hematopathology Laboratory, ACTREC, Tata Memorial Centre, Homi Bhabha National Institute, Mumbai, India
| | - Sweta Rajpal
- Hematopathology Laboratory, ACTREC, Tata Memorial Centre, Homi Bhabha National Institute, Mumbai, India
| | - Nikhil V Patkar
- Hematopathology Laboratory, ACTREC, Tata Memorial Centre, Homi Bhabha National Institute, Mumbai, India
| | - Bhausaheb Bagal
- Department of Medical Oncology, Tata Memorial Centre, Homi Bhabha National Institute, Mumbai, India
| | - Hasmukh Jain
- Department of Medical Oncology, Tata Memorial Centre, Homi Bhabha National Institute, Mumbai, India
| | - Manju Sengar
- Department of Medical Oncology, Tata Memorial Centre, Homi Bhabha National Institute, Mumbai, India
| | - Syed Khizer Hasan
- Cell and Tumor Biology Group, ACTREC, Tata Memorial Centre, Homi Bhabha National Institute, Mumbai, India
| | - Navin Khattry
- Department of Medical Oncology, Tata Memorial Centre, Homi Bhabha National Institute, Mumbai, India
| | - Papagudi G Subramanian
- Hematopathology Laboratory, ACTREC, Tata Memorial Centre, Homi Bhabha National Institute, Mumbai, India
| | - Sumeet Gujral
- Hematopathology Laboratory, ACTREC, Tata Memorial Centre, Homi Bhabha National Institute, Mumbai, India
| | - Prashant R Tembhare
- Hematopathology Laboratory, ACTREC, Tata Memorial Centre, Homi Bhabha National Institute, Mumbai, India
| |
Collapse
|
22
|
Abstract
Acute leukemia (AL) is a hematological malignancy, and the prognosis of most AL patients hasn’t improved significantly, particularly for relapsed or refractory (R/R) AL. Therefore, new treatments for R/R adult acute myeloid leukemia (AML) and acute lymphoblastic leukemia (ALL) are urgently necessary. Novel developments have been made in AL treatment, including target and immune therapies. CD38 is one of the targets due to its high expression in many hematological malignancies, including multiple myeloma, ALL and a subset of AML. Consequently, targeting CD38 therapies, including CD38 monoclonal antibodies (mAbs), bispecific antibodies, and CAR-T cell therapy, exhibit promising efficacy in treating multiple myeloma without significant toxicity and are being explored in other hematological malignancies and nonhematological diseases. Herein, this review focuses on targeting CD38 therapies in ALL and AML, which demonstrate sound antileukemic effects in acute leukemia and are expected to become effective treatment methods.
Collapse
|
23
|
Molecular Determinants Underlying the Anti-Cancer Efficacy of CD38 Monoclonal Antibodies in Hematological Malignancies. Biomolecules 2022; 12:biom12091261. [PMID: 36139103 PMCID: PMC9496523 DOI: 10.3390/biom12091261] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2022] [Revised: 08/31/2022] [Accepted: 09/02/2022] [Indexed: 11/17/2022] Open
Abstract
CD38 was first discovered as a T-cell antigen and has since been found ubiquitously expressed in various hematopoietic cells, including plasma cells, NK cells, B cells, and granulocytes. More importantly, CD38 expression levels on malignant hematopoietic cells are significantly higher than counterpart healthy cells, thus presenting itself as a promising therapeutic target. In fact, for many aggressive hematological cancers, including CLL, DLBCL, T-ALL, and NKTL, CD38 expression is significantly associated with poorer prognosis and a hyperproliferative or metastatic phenotype. Studies have shown that, beyond being a biomarker, CD38 functionally mediates dysregulated survival, adhesion, and migration signaling pathways, as well as promotes an immunosuppressive microenvironment conducive for tumors to thrive. Thus, targeting CD38 is a rational approach to overcoming these malignancies. However, clinical trials have surprisingly shown that daratumumab monotherapy has not been very effective in these other blood malignancies. Furthermore, extensive use of daratumumab in MM is giving rise to a subset of patients now refractory to daratumumab treatment. Thus, it is important to consider factors modulating the determinants of response to CD38 targeting across different blood malignancies, encompassing both the transcriptional and post-transcriptional levels so that we can diversify the strategy to enhance daratumumab therapeutic efficacy, which can ultimately improve patient outcomes.
Collapse
|
24
|
Newman H, Teachey DT. A Bright Horizon: Immunotherapy for Pediatric T-Cell Malignancies. Int J Mol Sci 2022; 23:8600. [PMID: 35955734 PMCID: PMC9369002 DOI: 10.3390/ijms23158600] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2022] [Revised: 07/27/2022] [Accepted: 08/01/2022] [Indexed: 02/04/2023] Open
Abstract
Immunotherapy has transformed the treatment of hematologic malignancies in the past two decades. The treatment of acute lymphoblastic leukemia (ALL), in particular, has been highly impacted by multiple novel immunotherapies. For pediatric patients with T-cell malignancies, translating immunotherapies has proved more challenging due to the complexities of fratricide, risk of product contamination with malignant cells, and concerns over T-cell aplasia. Despite these hurdles, many creative and promising strategies are on the horizon. We review challenges in the development of immunotherapy for T-cell malignancies, strategies to overcome these challenges, as well as therapies currently being investigated and starting to reach the clinic. Immunotherapy will hopefully successfully treat patients with relapsed and refractory T-cell malignancies and may someday be incorporated in up-front protocols in order to prevent relapses.
Collapse
Affiliation(s)
- Haley Newman
- Division of Oncology, The Children’s Hospital of Philadelphia, Philadelphia, PA 19104, USA;
- Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - David T. Teachey
- Division of Oncology, The Children’s Hospital of Philadelphia, Philadelphia, PA 19104, USA;
- Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| |
Collapse
|
25
|
Bommannan K, Arumugam JR, Radhakrishnan V, Kalaiyarasi JP, Karunakaran P, Mehra N, Sagar TG, Sundersingh S. Relevance of flow cytometry categorization and end-of-induction measurable residual disease assessment in pediatric and adult T-lymphoblastic leukemia patients. Blood Res 2022; 57:175-196. [PMID: 35880498 PMCID: PMC9492521 DOI: 10.5045/br.2022.2022104] [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: 05/16/2022] [Revised: 06/15/2022] [Accepted: 06/21/2022] [Indexed: 11/24/2022] Open
Abstract
Background T-lymphoblastic leukemia (T-ALL) patients expressing myeloid/stem cell antigens are classified as early T-cell precursor lymphoblastic leukemia (ETP-ALL) or near-ETP-ALL. Methods Clinico-laboratory profiles, flow cytometric end-of-induction measurable residual disease (EOI-MRD), and survival of treatment naïve T-ALL patients were analyzed according to their immunophenotypic subtypes. Results Among 81 consecutive T-ALL patients diagnosed, 21% (N=17) were ETP-ALL and 19% (N=15) were near-ETP-ALL. EOI-MRD was detectable in 39% of the 59 samples tested (31.6% of pediatric samples and 52.4% of adult samples). The frequency of EOI-MRD positivity was significantly higher among ETP-ALL (75%, P=0.001) and near-ETP-ALL (71%, P=0.009) patients compared to that in conventional-T-ALL (con-T-ALL) patients (22.5%). CD8 (P=0.046) and CD38 (P=0.046) expressions were significantly upregulated in the EOI blasts of con-T-ALL and ETP-ALL samples, respectively. The 2-year rates of overall (OS), relapse-free (RFS), and event-free survival (EFS) among the T-ALL patients (pediatric vs. adult) was 79.5% vs. 39.8% (P<0.001), 84.3% vs. 60.4% (P=0.026), and 80.3% vs. 38% (P<0.001), respectively. Univariate analysis revealed that 2-year EFS and RFS of pediatric T-ALL patients was independent of T-ALL subtype and was influenced only by EOI-MRD status. However, 2-year OS, RFS, and EFS among adult T-ALL patients were EOI-MRD independent and influenced only by the near-ETP-ALL phenotype. Conclusion Two-year survival among pediatric and adult T-ALL patients is attributed to EOI-MRD status and near-ETP-ALL phenotype, respectively.
Collapse
Affiliation(s)
- Karthik Bommannan
- Departments of Oncopathology, Cancer Institute (W.I.A.), Adyar, India
| | | | | | | | | | - Nikita Mehra
- Departments of Medical Oncology, Cancer Institute (W.I.A.), Adyar, India
| | - Tenali Gnana Sagar
- Departments of Medical Oncology, Cancer Institute (W.I.A.), Adyar, India
| | | |
Collapse
|
26
|
Combining daratumumab with CD47 blockade prolongs survival in preclinical models of pediatric T-ALL. Blood 2022; 140:45-57. [PMID: 35452517 DOI: 10.1182/blood.2021014485] [Citation(s) in RCA: 23] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2021] [Accepted: 04/18/2022] [Indexed: 11/20/2022] Open
Abstract
Acute lymphoblastic leukemia (ALL) is the most common malignant disease affecting children. Although therapy strategies improved, T-cell acute lymphoblastic leukemia (T-ALL) relapse is associated with chemoresistance and a poor prognosis. One strategy to overcome this obstacle is the application of monoclonal antibodies. Here, we show that leukemic cells from T-ALL patients express surface CD38 and CD47, both attractive targets for antibody therapy. We therefore investigated the commercially available CD38 antibody daratumumab (Dara) in combination with a proprietary modified CD47 antibody (Hu5F9-IgG2σ) in vitro and in vivo. Compared to single treatments, this combination significantly increased in vitro antibody-dependent cellular phagocytosis (ADCP) in T-ALL cell lines as well as in random de novo and in relapsed/refractory T-ALL patient derived xenograft (PDX) samples. Similarly, enhanced ADCP was observed when combining Dara with pharmacological inhibition of CD47 interactions using a glutaminyl cyclase inhibitor. Phase II-like preclinical in vivo trials using T-ALL PDX samples in experimental minimal residual disease like (MRD-like) and overt leukemia models revealed a high anti-leukemic efficacy of CD47 blockade alone. However, T-ALL xenograft mice subjected to chemotherapy first (post-chemo MRD) and subsequently co-treated with Dara and Hu5F9-IgG2σ displayed significantly reduced bone marrow infiltration as compared to single treatments. In relapsed and highly refractory T-ALL PDX combined treatment with Dara and Hu5F9-IgG2σ was required to substantially prolong survival as compared to single treatments. These findings suggest that combining CD47 blockade with Dara is a promising therapy for T-ALL, especially for relapsed/refractory disease harbouring a dismal prognosis in patients.
Collapse
|
27
|
NT5E gene and CD38 protein as potential prognostic biomarkers for childhood B-acute lymphoblastic leukemia. Purinergic Signal 2022; 18:211-222. [PMID: 35235138 DOI: 10.1007/s11302-022-09841-x] [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: 09/21/2021] [Accepted: 01/04/2022] [Indexed: 12/21/2022] Open
Abstract
The risk stratification of B-acute lymphoblastic leukemia (B-ALL) is based on clinical and biological factors. However, B-ALL has significant biological and clinical heterogeneity and 50% of B-ALL patients do not have defined prognostic markers. In this sense, the identification of new prognostic biomarkers is necessary. Considering different cohorts of childhood B-ALL patients, gene (DPP4/CD38/ENTPD1/NT5E) and protein (CD38/CD39/CD73) expressions of ectonucleotidases were analyzed in silico and ex vivo and the association with prognosis was established. In univariate analyses, expression of NT5E was significantly associated with worse progression-free survival (PFS) in bone marrow (BM) samples. In multivariate analyses, Kaplan-Meier analysis, and log-rank test, higher NT5E expression predicted unfavorable PFS in BM samples. Considering minimal residual disease (MRD), higher levels of cellularity were associated with the high NT5E expression at day 8 of induction therapy. In addition, we observed that white blood cells (WBC) of childhood B-ALL patients had more CD38 compared to the same cell population of healthy donors (HD). In fact, MRD > 0.1% patients had higher CD38 protein expression on WBC in comparison to HD. Noteworthy, we observed higher CD38 expression on WBC than blasts in MRD > 0.1% patients. We suggest that NT5E gene and CD38 protein expression, of the ectonucleotidases family, could provide interesting prognostic biomarkers for childhood B-ALL.
Collapse
|
28
|
Boissel N, Chevallier P, Doronin V, Griskevicius L, Maschan A, McCloskey J, Rambaldi A, Rossi G, Sokolov A, Wartiovaara-Kautto U, Oprea C, Abbadessa G, Gosselin A, Macé S, Thomas X. Isatuximab monotherapy in patients with refractory T-acute lymphoblastic leukemia or T-lymphoblastic lymphoma: Phase 2 study. Cancer Med 2022; 11:1292-1298. [PMID: 35106962 PMCID: PMC8894690 DOI: 10.1002/cam4.4478] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2021] [Revised: 10/04/2021] [Accepted: 10/26/2021] [Indexed: 12/19/2022] Open
Abstract
The poor prognosis of acute T‐cell lymphoblastic leukemia (T‐ALL) and T‐cell lymphoblastic lymphoma (T‐LBL) in older adults and patients with relapsed/refractory illness is an unmet clinical need, as there is no defined standard of care and there are few treatment options. Abnormally elevated CD38 expression in T‐ALL and T‐LBL is associated with tumor expansion and disease development, making CD38 a potential target for anti‐T‐ALL and T‐LBL treatment. Isatuximab is a monoclonal antibody that binds to a specific epitope on CD38. The purpose of the study was to assess the efficacy and safety of isatuximab monotherapy in a phase 2, multicenter, one‐arm, open‐label study in patients with relapsed or refractory T‐ALL or T‐LBL (Clinical Trials.gov identifier NCT02999633). The primary endpoint was to assess the efficacy of isatuximab by overall response rate (ORR). An interim analysis based on the efficacy and safety of isatuximab in the first 19 patients enrolled was scheduled, however only 14 patients were enrolled in the study. No patient achieved complete response (CR) or CR with incomplete peripheral recovery. Most patients (11 [78.6%]) developed progressive disease and had progressive disease as their best response. A total of 10 (71.4%) patients had treatment emergent adverse events considered treatment‐related, with infusion reactions as the most frequent drug‐related TEAE, occurring in 8 (57.1%) patients. Despite the low efficacy of isatuximab in the current study, it is likely that the use of immunotherapy medication in T‐ALL will be expanded through logically targeted approaches, together with advances in the design of T‐cell therapy and clinical experience and will provide restorative options beyond chemotherapy and targeted treatments.
Collapse
Affiliation(s)
- Nicolas Boissel
- Department of Adult Hematology, Saint-Louis Hospital, Institut de Recherche Saint-Louis, University of Paris, Paris, France
| | | | - Vadim Doronin
- Municipal Clinical Hospital, Moscow, Russian Federation
| | - Laimonas Griskevicius
- Hematology, Oncology and Transfusion Medicine Center, Vilnius University Hospital Santaros Klinikos, Vilnius University, Vilnius, Lithuania
| | - Alexey Maschan
- Federal Research Center for Pediatric Hematology, Oncology, and Immunology, Moscow, Russian Federation
| | - James McCloskey
- Division of Leukemia, John Theurer Cancer Center at Hackensack University Medical Center, Hackensack, New Jersey, USA
| | - Alessandro Rambaldi
- Università Statale di Milano, Milan and Azienda Socio Sanitaria Territoriale, Bergamo, Italy
| | - Giuseppe Rossi
- Divisione di Ematologia, ASST Spedali Civili, Brescia, Italy
| | - Andrey Sokolov
- National Research Center for Hematology of the Russian Ministry of Health, Moscow, Russian Federation
| | - Ulla Wartiovaara-Kautto
- Comprehensive Cancer Center, Department of Hematology, Helsinki University Hospital, Helsinki, Finland
| | | | | | | | | | - Xavier Thomas
- Hematology Department, Hospices Civils de Lyon, Lyon-Sud University Hospital, Pierre Bénite, France
| |
Collapse
|
29
|
Sin CF, Man PHM. Early T-Cell Precursor Acute Lymphoblastic Leukemia: Diagnosis, Updates in Molecular Pathogenesis, Management, and Novel Therapies. Front Oncol 2021; 11:750789. [PMID: 34912707 PMCID: PMC8666570 DOI: 10.3389/fonc.2021.750789] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2021] [Accepted: 11/04/2021] [Indexed: 12/26/2022] Open
Abstract
Early T-cell precursor acute lymphoblastic leukemia (ETP-ALL) is a distinct subtype of T lymphoblastic leukemia (T-ALL) identified in 2009, due to its unique immunophenotypic and genomic profile. The outcome of patients was poor in earlier studies, and they were prone to have induction failure, with more frequent relapse/refractory disease. Recent advances had been made in discoveries of genetic aberrations and molecular pathogenesis of ETP-ALL. However, the diagnosis and management of ETP-ALL is still challenging. There are limited choices of novel therapies so far. In this review article, it highlighted the diagnostic issue of ETP-ALL, pitfall in diagnosis, and strategy of accurate diagnosis. The review also summarized current understanding of molecular mechanism of leukemogenesis. The emerging role of risk-adapted therapy and allogenic stem cell transplant in optimizing the outcome of patients with ETP-ALL was discussed. Finally, some potential novel therapies were proposed based on the current understanding of molecular pathogenesis.
Collapse
Affiliation(s)
- Chun-fung Sin
- Department of Pathology, University of Hong Kong, Hong Kong, Hong Kong SAR, China
| | | |
Collapse
|
30
|
Multiparametric Flow Cytometry for MRD Monitoring in Hematologic Malignancies: Clinical Applications and New Challenges. Cancers (Basel) 2021; 13:cancers13184582. [PMID: 34572809 PMCID: PMC8470441 DOI: 10.3390/cancers13184582] [Citation(s) in RCA: 31] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2021] [Revised: 09/05/2021] [Accepted: 09/08/2021] [Indexed: 02/06/2023] Open
Abstract
Simple Summary In hematologic cancers, Minimal Residual Disease (MRD) monitoring, using either molecular (PCR) or immunophenotypic (MFC) diagnostics, allows the identification of rare cancer cells, readily detectable either in the bone marrow or in the peripheral blood at very low levels, far below the limit of classic microscopy. In this paper, we outlined the state-of-the-art of MFC-based MRD detection in different hematologic settings, highlighting main recommendations and new challenges for using such method in patients with acute leukemias or chronic hematologic neoplasms. The combination of new molecular technologies with advanced flow cytometry is progressively allowing clinicians to design a personalized therapeutic path, proportionate to the biological aggressiveness of the disease, in particular by using novel immunotherapies, in view of a modern decision-making process, based on precision medicine. Abstract Along with the evolution of immunophenotypic and molecular diagnostics, the assessment of Minimal Residual Disease (MRD) has progressively become a keystone in the clinical management of hematologic malignancies, enabling valuable post-therapy risk stratifications and guiding risk-adapted therapeutic approaches. However, specific prognostic values of MRD in different hematological settings, as well as its appropriate clinical uses (basically, when to measure it and how to deal with different MRD levels), still need further investigations, aiming to improve standardization and harmonization of MRD monitoring protocols and MRD-driven therapeutic strategies. Currently, MRD measurement in hematological neoplasms with bone marrow involvement is based on advanced highly sensitive methods, able to detect either specific genetic abnormalities (by PCR-based techniques and next-generation sequencing) or tumor-associated immunophenotypic profiles (by multiparametric flow cytometry, MFC). In this review, we focus on the growing clinical role for MFC-MRD diagnostics in hematological malignancies—from acute myeloid and lymphoblastic leukemias (AML, B-ALL and T-ALL) to chronic lymphocytic leukemia (CLL) and multiple myeloma (MM)—providing a comparative overview on technical aspects, clinical implications, advantages and pitfalls of MFC-MRD monitoring in different clinical settings.
Collapse
|
31
|
Salvaris R, Fedele PL. Targeted Therapy in Acute Lymphoblastic Leukaemia. J Pers Med 2021; 11:715. [PMID: 34442359 PMCID: PMC8398498 DOI: 10.3390/jpm11080715] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2021] [Accepted: 07/21/2021] [Indexed: 11/26/2022] Open
Abstract
The last decade has seen a significant leap in our understanding of the wide range of genetic lesions underpinning acute lymphoblastic leukaemia (ALL). Next generation sequencing has led to the identification of driver mutations with significant implications on prognosis and has defined entities such as BCR-ABL-like ALL, where targeted therapies such as tyrosine kinase inhibitors (TKIs) and JAK inhibitors may play a role in its treatment. In Philadelphia positive ALL, the introduction of TKIs into frontline treatment regimens has already transformed patient outcomes. In B-ALL, agents targeting surface receptors CD19, CD20 and CD22, including monoclonal antibodies, bispecific T cell engagers, antibody drug conjugates and chimeric antigen receptor (CAR) T cells, have shown significant activity but come with unique toxicities and have implications for how treatment is sequenced. Advances in T-ALL have lagged behind those seen in B-ALL. However, agents such as nelarabine, bortezomib and CAR T cell therapy targeting T cell antigens have been examined with promising results seen. As our understanding of disease biology in ALL grows, as does our ability to target pathways such as apoptosis, through BH3 mimetics, chemokines and epigenetic regulators. This review aims to highlight a range of available and emerging targeted therapeutics in ALL, to explore their mechanisms of action and to discuss the current evidence for their use.
Collapse
Affiliation(s)
- Ross Salvaris
- Department of Clinical Haematology, Monash Health, Clayton 3168, Australia;
- School of Clinical Sciences at Monash Health, Monash University, Clayton 3168, Australia
| | - Pasquale Luke Fedele
- Department of Clinical Haematology, Monash Health, Clayton 3168, Australia;
- School of Clinical Sciences at Monash Health, Monash University, Clayton 3168, Australia
| |
Collapse
|
32
|
Advances in immunotherapeutic targets for childhood cancers: A focus on glypican-2 and B7-H3. Pharmacol Ther 2021; 223:107892. [PMID: 33992682 DOI: 10.1016/j.pharmthera.2021.107892] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2021] [Revised: 05/05/2021] [Accepted: 05/10/2021] [Indexed: 12/16/2022]
Abstract
Cancer immunotherapies have revolutionized how we can treat adult malignancies and are being translated to pediatric oncology. Chimeric antigen receptor T-cell therapy and bispecific antibodies targeting CD19 have shown success for the treatment of pediatric patients with B-cell acute lymphoblastic leukemia. Anti-GD2 monoclonal antibody has demonstrated efficacy in neuroblastoma. In this review, we summarize the immunotherapeutic agents that have been approved for treating childhood cancers and provide an updated review of molecules expressed by pediatric cancers that are under study or are emerging candidates for future immunotherapies. Advances in our knowledge of tumor immunology and in genome profiling of cancers has led to the identification of new tumor-specific/associated antigens. While cell surface antigens are normally targeted in a major histocompatibility complex (MHC)-independent manner using antibody-based therapies, intracellular antigens are normally targeted with MHC-dependent T cell therapies. Glypican 2 (GPC2) and B7-H3 (CD276) are two cell surface antigens that are expressed by a variety of pediatric tumors such as neuroblastoma and potentially can have a positive impact on the treatment of pediatric cancers in the clinic.
Collapse
|
33
|
Facts and Challenges in Immunotherapy for T-Cell Acute Lymphoblastic Leukemia. Int J Mol Sci 2020; 21:ijms21207685. [PMID: 33081391 PMCID: PMC7589289 DOI: 10.3390/ijms21207685] [Citation(s) in RCA: 36] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2020] [Revised: 10/13/2020] [Accepted: 10/14/2020] [Indexed: 12/12/2022] Open
Abstract
T-cell acute lymphoblastic leukemia (T-ALL), a T-cell malignant disease that mainly affects children, is still a medical challenge, especially for refractory patients for whom therapeutic options are scarce. Recent advances in immunotherapy for B-cell malignancies based on increasingly efficacious monoclonal antibodies (mAbs) and chimeric antigen receptors (CARs) have been encouraging for non-responding or relapsing patients suffering from other aggressive cancers like T-ALL. However, secondary life-threatening T-cell immunodeficiency due to shared expression of targeted antigens by healthy and malignant T cells is a main drawback of mAb—or CAR-based immunotherapies for T-ALL and other T-cell malignancies. This review provides a comprehensive update on the different immunotherapeutic strategies that are being currently applied to T-ALL. We highlight recent progress on the identification of new potential targets showing promising preclinical results and discuss current challenges and opportunities for developing novel safe and efficacious immunotherapies for T-ALL.
Collapse
|