1
|
Schoenfeld K, Habermann J, Wendel P, Harwardt J, Ullrich E, Kolmar H. T cell receptor-directed antibody-drug conjugates for the treatment of T cell-derived cancers. MOLECULAR THERAPY. ONCOLOGY 2024; 32:200850. [PMID: 39176070 PMCID: PMC11338945 DOI: 10.1016/j.omton.2024.200850] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/30/2024] [Revised: 07/02/2024] [Accepted: 07/16/2024] [Indexed: 08/24/2024]
Abstract
T cell-derived cancers are hallmarked by heterogeneity, aggressiveness, and poor clinical outcomes. Available targeted therapies are severely limited due to a lack of target antigens that allow discrimination of malignant from healthy T cells. Here, we report a novel approach for the treatment of T cell diseases based on targeting the clonally rearranged T cell receptor displayed by the cancerous T cell population. As a proof of concept, we identified an antibody with unique specificity toward a distinct T cell receptor (TCR) and developed antibody-drug conjugates, precisely recognizing and eliminating target T cells while preserving overall T cell repertoire integrity and cellular immunity. Our anti-TCR antibody-drug conjugates demonstrated effective receptor-mediated cell internalization, associated with induction of cancer cell death with strong signs of apoptosis. Furthermore, cell proliferation-inhibiting bystander effects observed on target-negative cells may contribute to the molecules' anti-tumor properties precluding potential tumor escape mechanisms. To our knowledge, this represents the first anti-TCR antibody-drug conjugate designed as custom-tailored immunotherapy for T cell-driven pathologies.
Collapse
Affiliation(s)
- Katrin Schoenfeld
- Institute for Organic Chemistry and Biochemistry, Technical University of Darmstadt, 64287 Darmstadt, Germany
| | - Jan Habermann
- Institute for Organic Chemistry and Biochemistry, Technical University of Darmstadt, 64287 Darmstadt, Germany
- Goethe University, Department of Pediatrics, Experimental Immunology and Cell Therapy, 60590 Frankfurt am Main, Germany
- Frankfurt Cancer Institute, Goethe University, 60596 Frankfurt am Main, Germany
| | - Philipp Wendel
- Institute for Organic Chemistry and Biochemistry, Technical University of Darmstadt, 64287 Darmstadt, Germany
- Goethe University, Department of Pediatrics, Experimental Immunology and Cell Therapy, 60590 Frankfurt am Main, Germany
- Frankfurt Cancer Institute, Goethe University, 60596 Frankfurt am Main, Germany
- German Cancer Consortium (DKTK), partner site Frankfurt/Mainz, 60590 Frankfurt am Main, Germany
- German Cancer Research Center (DKFZ), 69120 Heidelberg, Germany
| | - Julia Harwardt
- Institute for Organic Chemistry and Biochemistry, Technical University of Darmstadt, 64287 Darmstadt, Germany
| | - Evelyn Ullrich
- Goethe University, Department of Pediatrics, Experimental Immunology and Cell Therapy, 60590 Frankfurt am Main, Germany
- Frankfurt Cancer Institute, Goethe University, 60596 Frankfurt am Main, Germany
- German Cancer Consortium (DKTK), partner site Frankfurt/Mainz, 60590 Frankfurt am Main, Germany
| | - Harald Kolmar
- Institute for Organic Chemistry and Biochemistry, Technical University of Darmstadt, 64287 Darmstadt, Germany
- Centre for Synthetic Biology, Technical University of Darmstadt, 64283 Darmstadt, Germany
| |
Collapse
|
2
|
Galante O, Bleier H, Levi I, Fuchs L, Almog Y, Shafat T. Outcomes of non-COVID-19 critically ill patients with hematologic malignancies a 10-year single-center retrospective analysis. Leuk Lymphoma 2024:1-8. [PMID: 39262397 DOI: 10.1080/10428194.2024.2401082] [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: 05/08/2024] [Revised: 08/18/2024] [Accepted: 08/31/2024] [Indexed: 09/13/2024]
Abstract
We report the outcomes of patients with haematological malignancies admitted to ICUs and define pre-ICU prognostic factors for in-hospital mortality. In a retrospective, single-center study, we included all patients with haematologic malignancies admitted to ICUs between 2009 and 2019. The primary outcome was in-hospital mortality. One hundred and forty-four patients with hematologic malignancies were admitted to ICUs during the study period. Fifteen (10.4%) were in remission, 36 (25.0%) were in remission after hematopoietic stem cell transplantation. Acute Leukemias and aggressive lymphomas were the most common diagnoses, occurring in 34.7%. The in-hospital mortality was 49%. The main predictors for in-hospital mortality were age >65 years, post allogeneic hematopoietic stem cell transplantation, non-remission, respiratory rate >22 bpm, bilirubin >2 mg/dl, PH< 7.35, and time from hospital admission to ICU transfer ≥3 days. In-hospital mortality of patients with hematologic malignancies admitted to ICU was 49%. We identified pre-ICU parameters that predict in-hospital mortality.
Collapse
Affiliation(s)
- Ori Galante
- Medical Intensive Care Unit, Soroka University Medical Center, Beer Sheva, Israel
- Faculty of Health Science, Ben Gurion University of the Negev, Beer Sheva, Israel
| | - Hamutal Bleier
- Faculty of Health Science, Ben Gurion University of the Negev, Beer Sheva, Israel
| | - Itai Levi
- Faculty of Health Science, Ben Gurion University of the Negev, Beer Sheva, Israel
- Hematology Institute, Soroka University Medical Center, Beer Sheva, Israel
| | - Lior Fuchs
- Medical Intensive Care Unit, Soroka University Medical Center, Beer Sheva, Israel
- Faculty of Health Science, Ben Gurion University of the Negev, Beer Sheva, Israel
| | - Yaniv Almog
- Medical Intensive Care Unit, Soroka University Medical Center, Beer Sheva, Israel
- Faculty of Health Science, Ben Gurion University of the Negev, Beer Sheva, Israel
| | - Tali Shafat
- Faculty of Health Science, Ben Gurion University of the Negev, Beer Sheva, Israel
- Infectious Diseases Institute, Soroka University Medical Center, Beer Sheva, Israel
- Clinical Research Center, Soroka University Medical Center, Beer Sheva, Israel
| |
Collapse
|
3
|
Kim J, Cho J, Lee MH, Yoon SE, Kim WS, Kim SJ. CAR T cells vs bispecific antibody as third- or later-line large B-cell lymphoma therapy: a meta-analysis. Blood 2024; 144:629-638. [PMID: 38696731 DOI: 10.1182/blood.2023023419] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2023] [Revised: 04/23/2024] [Accepted: 04/23/2024] [Indexed: 05/04/2024] Open
Abstract
ABSTRACT This meta-analysis evaluates the efficacy and safety of chimeric antigen receptor (CAR) T-cell therapy and bispecific antibodies for relapsed/refractory diffuse large B-cell lymphoma (R/R DLBCL). We searched MEDLINE, Embase, and Cochrane databases until July 2023 for trials assessing CAR T-cell therapies and CD20×CD3 bispecific antibodies as third or subsequent lines in R/R DLBCL. Random-effects models estimated the complete response (CR) rate and secondary outcomes, with meta-regressions adjusting for relevant covariates. Sixteen studies comprising 1347 patients were included in the pooled analysis. The pooled CR rate for bispecific antibodies was 0.36 (95% confidence interval [CI], 0.29-0.43), compared with 0.51 (95% CI, 0.46-0.56) for CAR T-cell therapy (P < .01). This superiority persisted when comparing the CAR T-cell-naive patients within the bispecific antibody group, with a CR rate of 0.37 (95% CI, 0.32-0.43). Multivariable meta-regression also revealed better efficacy of CAR T cells with adjustment for the proportion of double-hit lymphoma. The pooled 1-year progression-free survival rate mirrored these findings (0.32 [95% CI, 0.26-0.38] vs 0.44 [95% CI, 0.41-0.48]; P < .01). For adverse events of grade ≥3, the bispecific antibody had incidences of 0.02 (95% CI, 0.01-0.04) for cytokine release syndrome, 0.01 (95% CI, 0.00-0.01) for neurotoxicity, and 0.10 (95% CI, 0.03-0.16) for infections. The CAR T cell had rates of 0.08 (95% CI, 0.03-0.12), 0.11 (95% CI, 0.06-0.17), and 0.17 (95% CI, 0.11-0.22), respectively, with significant differences observed in the first 2 categories. In summary, CAR T-cell therapy outperformed bispecific antibody in achieving higher CR rates, although with an increase in severe adverse events.
Collapse
Affiliation(s)
- Jinchul Kim
- Department of Hematology-Oncology, Inha University College of Medicine and Hospital, Incheon, Korea
| | - Jinhyun Cho
- Department of Hematology-Oncology, Inha University College of Medicine and Hospital, Incheon, Korea
| | - Moon Hee Lee
- Department of Hematology-Oncology, Inha University College of Medicine and Hospital, Incheon, Korea
| | - Sang Eun Yoon
- Division of Hematology-Oncology, Department of Medicine, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Korea
| | - Won Seog Kim
- Division of Hematology-Oncology, Department of Medicine, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Korea
- Department of Health Sciences and Technology, Samsung Advanced Institute for Health Sciences and Technology, Sungkyunkwan University School of Medicine, Seoul, Korea
| | - Seok Jin Kim
- Division of Hematology-Oncology, Department of Medicine, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Korea
- Department of Health Sciences and Technology, Samsung Advanced Institute for Health Sciences and Technology, Sungkyunkwan University School of Medicine, Seoul, Korea
| |
Collapse
|
4
|
Isavand P, Aghamiri SS, Amin R. Applications of Multimodal Artificial Intelligence in Non-Hodgkin Lymphoma B Cells. Biomedicines 2024; 12:1753. [PMID: 39200217 PMCID: PMC11351272 DOI: 10.3390/biomedicines12081753] [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: 06/25/2024] [Revised: 07/22/2024] [Accepted: 08/01/2024] [Indexed: 09/02/2024] Open
Abstract
Given advancements in large-scale data and AI, integrating multimodal artificial intelligence into cancer research can enhance our understanding of tumor behavior by simultaneously processing diverse biomedical data types. In this review, we explore the potential of multimodal AI in comprehending B-cell non-Hodgkin lymphomas (B-NHLs). B-cell non-Hodgkin lymphomas (B-NHLs) represent a particular challenge in oncology due to tumor heterogeneity and the intricate ecosystem in which tumors develop. These complexities complicate diagnosis, prognosis, and therapy response, emphasizing the need to use sophisticated approaches to enhance personalized treatment strategies for better patient outcomes. Therefore, multimodal AI can be leveraged to synthesize critical information from available biomedical data such as clinical record, imaging, pathology and omics data, to picture the whole tumor. In this review, we first define various types of modalities, multimodal AI frameworks, and several applications in precision medicine. Then, we provide several examples of its usage in B-NHLs, for analyzing the complexity of the ecosystem, identifying immune biomarkers, optimizing therapy strategy, and its clinical applications. Lastly, we address the limitations and future directions of multimodal AI, highlighting the need to overcome these challenges for better clinical practice and application in healthcare.
Collapse
Affiliation(s)
- Pouria Isavand
- Department of Radiology, School of Medicine, Zanjan University of Medical Sciences, Zanjan 4513956184, Iran
| | | | - Rada Amin
- Department of Biochemistry, University of Nebraska, Lincoln, NE 68503, USA
| |
Collapse
|
5
|
Olejarz W, Sadowski K, Szulczyk D, Basak G. Advancements in Personalized CAR-T Therapy: Comprehensive Overview of Biomarkers and Therapeutic Targets in Hematological Malignancies. Int J Mol Sci 2024; 25:7743. [PMID: 39062986 PMCID: PMC11276786 DOI: 10.3390/ijms25147743] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2024] [Revised: 07/12/2024] [Accepted: 07/12/2024] [Indexed: 07/28/2024] Open
Abstract
Chimeric antigen receptor T-cell (CAR-T) therapy is a novel anticancer therapy using autologous or allogeneic T-cells. To date, six CAR-T therapies for specific B-cell acute lymphoblastic leukemia (B-ALL), non-Hodgkin lymphomas (NHL), and multiple myeloma (MM) have been approved by the Food and Drug Administration (FDA). Significant barriers to the effectiveness of CAR-T therapy include cytokine release syndrome (CRS), neurotoxicity in the case of Allogeneic Stem Cell Transplantation (Allo-SCT) graft-versus-host-disease (GVHD), antigen escape, modest antitumor activity, restricted trafficking, limited persistence, the immunosuppressive microenvironment, and senescence and exhaustion of CAR-Ts. Furthermore, cancer drug resistance remains a major problem in clinical practice. CAR-T therapy, in combination with checkpoint blockades and bispecific T-cell engagers (BiTEs) or other drugs, appears to be an appealing anticancer strategy. Many of these agents have shown impressive results, combining efficacy with tolerability. Biomarkers like extracellular vesicles (EVs), cell-free DNA (cfDNA), circulating tumor (ctDNA) and miRNAs may play an important role in toxicity, relapse assessment, and efficacy prediction, and can be implicated in clinical applications of CAR-T therapy and in establishing safe and efficacious personalized medicine. However, further research is required to fully comprehend the particular side effects of immunomodulation, to ascertain the best order and combination of this medication with conventional chemotherapy and targeted therapies, and to find reliable predictive biomarkers.
Collapse
Affiliation(s)
- Wioletta Olejarz
- Department of Biochemistry and Pharmacogenomics, Faculty of Pharmacy, Medical University of Warsaw, 02-097 Warsaw, Poland;
- Centre for Preclinical Research, Medical University of Warsaw, 02-097 Warsaw, Poland
| | - Karol Sadowski
- Department of Biochemistry and Pharmacogenomics, Faculty of Pharmacy, Medical University of Warsaw, 02-097 Warsaw, Poland;
- Centre for Preclinical Research, Medical University of Warsaw, 02-097 Warsaw, Poland
- Department of Hematology, Transplantation and Internal Medicine, Medical University of Warsaw, 02-097 Warsaw, Poland;
| | - Daniel Szulczyk
- Chair and Department of Biochemistry, The Medical University of Warsaw, 02-097 Warsaw, Poland;
| | - Grzegorz Basak
- Department of Hematology, Transplantation and Internal Medicine, Medical University of Warsaw, 02-097 Warsaw, Poland;
| |
Collapse
|
6
|
Fu J, Hao Z. The causality between gut microbiota and non-Hodgkin lymphoma: a two-sample bidirectional Mendelian randomization study. Front Microbiol 2024; 15:1403825. [PMID: 38860220 PMCID: PMC11163074 DOI: 10.3389/fmicb.2024.1403825] [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: 03/20/2024] [Accepted: 05/10/2024] [Indexed: 06/12/2024] Open
Abstract
Background Studies have indicated an association between gut microbiota (GM) and non-Hodgkin lymphoma (NHL). However, the causality between GM and NHL remains unclear. This study aims to investigate the causality between GM and NHL using Mendelian randomization (MR). Methods Data on GM is sourced from the MiBioGen consortium, while data on NHL and its subtypes is sourced from the FinnGen consortium R10 version. Inverse variance weighted (IVW) was employed for the primary MR analysis method, with methods such as Bayesian weighted Mendelian randomisation (BWMR) as an adjunct. Sensitivity analyses were conducted using Cochran's Q test, MR-Egger regression, MR-PRESSO, and the "Leave-one-out" method. Results The MR results showed that there is a causality between 27 GMs and NHL. Among them, 20 were negatively associated (OR < 1), and 7 were positively associated (OR > 1) with the corresponding diseases. All 27 MR results passed sensitivity tests, and there was no reverse causal association. Conclusion By demonstrating a causal link between GM and NHL, this research offers novel ideas to prevent, monitor, and cure NHL later.
Collapse
Affiliation(s)
- Jinjie Fu
- Graduate School, Tianjin University of Traditional Chinese Medicine, Tianjin, China
- College of Traditional Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Zheng Hao
- College of Traditional Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, China
- Tianjin Key Laboratory of Modern Chinese Medicine Theory of Innovation and Application, Tianjin, China
- Guo Aichun Institute of Medical History and Literature, Tianjin University of Traditional Chinese Medicine, Tianjin, China
| |
Collapse
|
7
|
Mandala E, Lafara K, Kokkinovasilis D, Kalafatis I, Koukoulitsa V, Katodritou E, Lafaras C. Applied Cardio-Oncology in Hematological Malignancies: A Narrative Review. Life (Basel) 2024; 14:524. [PMID: 38672794 PMCID: PMC11050930 DOI: 10.3390/life14040524] [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: 03/19/2024] [Revised: 04/12/2024] [Accepted: 04/15/2024] [Indexed: 04/28/2024] Open
Abstract
Applied cardio-oncology in hematological malignancies refers to the integration of cardiovascular care and management for patients with blood cancer, particularly leukemia, lymphoma, and multiple myeloma. Hematological cancer therapy-related cardiotoxicity deals with the most common cardiovascular complications of conventional chemotherapy, targeted therapy, immunotherapy, chimeric antigen receptor T (CAR-T) cell and tumor-infiltrating lymphocyte therapies, bispecific antibodies, and hematopoietic stem cell transplantation. This narrative review focuses on hematological cancer-therapy-related cardiotoxicity's definition, risk stratification, multimodality imaging, and use of cardiac biomarkers to detect clinical and/or subclinical myocardial dysfunction and electrical instability. Moreover, the most common cardiotoxic profiles of the main drugs and/or therapeutic interventions in patients with hematological malignancies are described thoroughly.
Collapse
Affiliation(s)
- Evdokia Mandala
- Division of Hematology, Forth Department of Medicine, School of Medicine, Aristotle University of Thessaloniki, 54642 Thessaloniki, Greece; (E.M.); (K.L.); (D.K.)
| | - Kyranna Lafara
- Division of Hematology, Forth Department of Medicine, School of Medicine, Aristotle University of Thessaloniki, 54642 Thessaloniki, Greece; (E.M.); (K.L.); (D.K.)
| | - Dimitrios Kokkinovasilis
- Division of Hematology, Forth Department of Medicine, School of Medicine, Aristotle University of Thessaloniki, 54642 Thessaloniki, Greece; (E.M.); (K.L.); (D.K.)
| | - Ioannis Kalafatis
- Cardiology-Oncology Unit, Theagenion Cancer Hospital, 54639 Thessaloniki, Greece; (I.K.); (V.K.)
| | - Vasiliki Koukoulitsa
- Cardiology-Oncology Unit, Theagenion Cancer Hospital, 54639 Thessaloniki, Greece; (I.K.); (V.K.)
| | - Eirini Katodritou
- Department of Hematology, Theagenion Cancer Hospital, 54639 Thessaloniki, Greece;
| | - Christos Lafaras
- Cardiology-Oncology Unit, Theagenion Cancer Hospital, 54639 Thessaloniki, Greece; (I.K.); (V.K.)
| |
Collapse
|
8
|
Liu S, Xu M, Zhong L, Tong X, Qian S. Recent Advances in Nanobiotechnology for the Treatment of Non-Hodgkin's Lymphoma. Mini Rev Med Chem 2024; 24:895-907. [PMID: 37724679 DOI: 10.2174/1389557523666230915103121] [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: 04/15/2023] [Revised: 06/20/2023] [Accepted: 07/25/2023] [Indexed: 09/21/2023]
Abstract
Lymphoma is the eighth most common type of cancer worldwide. Currently, lymphoma is mainly classified into two main groups: Hodgkin's lymphoma (HL) and non-Hodgkin's lymphoma (NHL), with NHL accounting for 80% to 90% of the cases. NHL is primarily divided into B, T, and natural killer (NK) cell lymphoma. Nanotechnology is developing rapidly and has made significant contributions to the field of medicine. This review summarizes the advancements of nanobiotechnology in recent years and its applications in the treatment of NHL, especially in diffuse large B cell lymphoma (DLBCL), primary central nervous system lymphoma (PCNSL), and follicular lymphoma (FL). The technologies discussed include clinical imaging, targeted drug delivery, photodynamic therapy (PDT), and thermodynamic therapy (TDT) for lymphoma. This review aims to provide a better understanding of the use of nanotechnology in the treatment of non-Hodgkin's lymphoma.
Collapse
Affiliation(s)
- Shuxian Liu
- Key Laboratory of Elemene Class Anti-Cancer Chinese Medicines, School of Pharmacy, Hangzhou Normal University, Hangzhou, Zhejiang, 311121, China
| | - Minghao Xu
- Key Laboratory of Elemene Class Anti-Cancer Chinese Medicines, School of Pharmacy, Hangzhou Normal University, Hangzhou, Zhejiang, 311121, China
| | - Lei Zhong
- Tongxiang Hospital of Traditional Chinese Medicine, Zhejiang, China
| | - Xiangmin Tong
- Zhejiang Provincial People's Hospital, Affiliated People's Hospital, Hangzhou Medical College, Hangzhou, 310014, China
| | - Suying Qian
- Department of Hematology and Oncology, Ningbo No. 2 Hospital, China
| |
Collapse
|
9
|
Soueidy C, Kourie HR. Updates in the Management of Primary Mediastinal B Cell Lymphoma. CLINICAL LYMPHOMA, MYELOMA & LEUKEMIA 2023; 23:866-873. [PMID: 37722943 DOI: 10.1016/j.clml.2023.08.014] [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: 06/08/2023] [Revised: 08/17/2023] [Accepted: 08/18/2023] [Indexed: 09/20/2023]
Abstract
Primary mediastinal B cell lymphoma (PMBCL) is considered a distinct pathology according to the WHO classification of lymphoid malignancies. Patients have a better prognosis after the addition of Rituximab to anthracycline-based chemotherapy. The role of consolidative radiotherapy is controversial after the approval of dose-adjusted R-EPOCH and the selection of patients to undergo radiotherapy is based on end-of-therapy PET CT. In the relapsed/refractory setting, new approved drugs and other under investigation have improved patient outcomes. This review summarizes the different treatment modalities in (PMBCL) in the frontline and the relapsed/refractory settings.
Collapse
Affiliation(s)
- Charbel Soueidy
- Hematology Oncology Department, Hotel Dieu de France Hospital, Beirut, Lebanon.
| | | |
Collapse
|
10
|
Walseng E, Wang B, Yang C, Patel P, Zhao C, Zhang H, Zhao P, Mazor Y. Conformation-selective rather than avidity-based binding to tumor associated antigen derived peptide-MHC enables targeting of WT1-pMHC low expressing cancer cells by anti-WT1-pMHC/CD3 T cell engagers. Front Immunol 2023; 14:1275304. [PMID: 38022650 PMCID: PMC10667733 DOI: 10.3389/fimmu.2023.1275304] [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: 08/09/2023] [Accepted: 10/25/2023] [Indexed: 12/01/2023] Open
Abstract
T cell engagers, a category of T cell-retargeting immunotherapy, are rapidly transforming clinical cancer care. However, the lack of tumor-specific targets poses a significant roadblock for broad adaptation of this therapeutic modality in many indications, often resulting in systemic on-target off-tumor toxicity. Though various tumor-derived intracellular mutations provide a massive pool of potential tumor-specific antigens, targeting them is extremely challenging, partly due to the low copy number of tumor associated antigen (TAA)-derived pMHC on tumor cell surface. Further, the interplay of binding geometry and format valency in relation to the capacity of a T cell engager to efficiently target low density cell-surface pMHC is not well understood. Using the Wilms' tumor 1 (WT1) oncoprotein as a proof-of-principle TAA, combined with an array of IgG-like T cell engager modalities that differ in their anti-TAA valency and binding geometry, we show that the ability to induce an immunological synapse formation, resulting in potent killing of WT1 positive cancer cell lines is primarily dependent on the distinct geometrical conformations between the Fab arms of anti-WT1-HLA-A*02:01 and anti-CD3. The augmented avidity conferred by the binding of two anti-WT1-HLA-A*02:01 Fab arms has only minimal influence on cell killing potency. These findings demonstrate the need for careful examination of key design parameters for the development of next-generation T cell engagers targeting low density TAA-pMHCs on tumor cells.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | - Yariv Mazor
- Biologics Engineering, Biopharmaceutical R&D, AstraZeneca, Gaithersburg, MD, United States
| |
Collapse
|
11
|
Tang L, Huang Z, Mei H, Hu Y. Immunotherapy in hematologic malignancies: achievements, challenges and future prospects. Signal Transduct Target Ther 2023; 8:306. [PMID: 37591844 PMCID: PMC10435569 DOI: 10.1038/s41392-023-01521-5] [Citation(s) in RCA: 23] [Impact Index Per Article: 23.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2022] [Revised: 05/31/2023] [Accepted: 06/04/2023] [Indexed: 08/19/2023] Open
Abstract
The immune-cell origin of hematologic malignancies provides a unique avenue for the understanding of both the mechanisms of immune responsiveness and immune escape, which has accelerated the progress of immunotherapy. Several categories of immunotherapies have been developed and are being further evaluated in clinical trials for the treatment of blood cancers, including stem cell transplantation, immune checkpoint inhibitors, antigen-targeted antibodies, antibody-drug conjugates, tumor vaccines, and adoptive cell therapies. These immunotherapies have shown the potential to induce long-term remission in refractory or relapsed patients and have led to a paradigm shift in cancer treatment with great clinical success. Different immunotherapeutic approaches have their advantages but also shortcomings that need to be addressed. To provide clinicians with timely information on these revolutionary therapeutic approaches, the comprehensive review provides historical perspectives on the applications and clinical considerations of the immunotherapy. Here, we first outline the recent advances that have been made in the understanding of the various categories of immunotherapies in the treatment of hematologic malignancies. We further discuss the specific mechanisms of action, summarize the clinical trials and outcomes of immunotherapies in hematologic malignancies, as well as the adverse effects and toxicity management and then provide novel insights into challenges and future directions.
Collapse
Affiliation(s)
- Lu Tang
- Institute of Hematology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, 430022, Wuhan, China
- Hubei Clinical Medical Center of Cell Therapy for Neoplastic Disease, 430022, Wuhan, China
- Key Laboratory of Biological Targeted Therapy, the Ministry of Education, 430022, Wuhan, China
| | - Zhongpei Huang
- Institute of Hematology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, 430022, Wuhan, China
- Hubei Clinical Medical Center of Cell Therapy for Neoplastic Disease, 430022, Wuhan, China
- Key Laboratory of Biological Targeted Therapy, the Ministry of Education, 430022, Wuhan, China
| | - Heng Mei
- Institute of Hematology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, 430022, Wuhan, China.
- Hubei Clinical Medical Center of Cell Therapy for Neoplastic Disease, 430022, Wuhan, China.
- Key Laboratory of Biological Targeted Therapy, the Ministry of Education, 430022, Wuhan, China.
- Hubei Key Laboratory of Biological Targeted Therapy, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, 430022, Wuhan, China.
| | - Yu Hu
- Institute of Hematology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, 430022, Wuhan, China.
- Hubei Clinical Medical Center of Cell Therapy for Neoplastic Disease, 430022, Wuhan, China.
- Key Laboratory of Biological Targeted Therapy, the Ministry of Education, 430022, Wuhan, China.
- Hubei Key Laboratory of Biological Targeted Therapy, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, 430022, Wuhan, China.
| |
Collapse
|
12
|
Liu X, Zhao J, Guo X, Song Y. CD20 × CD3 bispecific antibodies for lymphoma therapy: latest updates from ASCO 2023 annual meeting. J Hematol Oncol 2023; 16:90. [PMID: 37537626 PMCID: PMC10401875 DOI: 10.1186/s13045-023-01488-4] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2023] [Accepted: 07/26/2023] [Indexed: 08/05/2023] Open
Abstract
Multiple bispecific antibodies (bsAbs) have been approved for cancer immunotherapy. Several CD20 × CD3 bsAbs have demonstrated significant anti-B-cell non-Hodgkin lymphoma (NHL) activity by engaging T cells to target CD20+ NHL cells in clinical trials. Mosunetuzumab, epcoritamab and glofitamab have been approved recently for B-cell NHL therapy. In this study, we summarized several latest reports on CD20 × CD3 bsAbs for the therapy of B-cell NHL from the ASCO 2023 annual meeting (ASCO2023).
Collapse
Affiliation(s)
- Xinyuan Liu
- Institute of Biomedical Informatics, Bioinformatics Center, Henan Provincial Engineering Center for Tumor Molecular Medicine, School of Basic Medical Sciences, Henan University, Kaifeng, 475004, Henan, China
| | - Juanjuan Zhao
- Department of Hematology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, Henan, China
| | - Xiangqian Guo
- Institute of Biomedical Informatics, Bioinformatics Center, Henan Provincial Engineering Center for Tumor Molecular Medicine, School of Basic Medical Sciences, Henan University, Kaifeng, 475004, Henan, China
| | - Yongping Song
- Department of Hematology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, Henan, China.
| |
Collapse
|
13
|
Meetze K, Mehta NK, Li B, Michaelson JS, Baeuerle PA. CLN-978, a novel half-life extended CD19/CD3/HSA-specific T cell-engaging antibody construct with potent activity against B-cell malignancies with low CD19 expression. J Immunother Cancer 2023; 11:e007398. [PMID: 37586770 PMCID: PMC10432633 DOI: 10.1136/jitc-2023-007398] [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] [Accepted: 07/30/2023] [Indexed: 08/18/2023] Open
Abstract
BACKGROUND Despite significant progress in the development of T cell-engaging therapies for various B-cell malignancies, a high medical need remains for the refractory disease setting, often characterized by suboptimal target levels. METHODS To address this issue, we have developed a 65-kDa multispecific antibody construct, CLN-978, with affinities tuned to optimize the killing of low-CD19 expressing tumor cells. CLN-978 bound to CD19 on B cells with picomolar affinity, and to CD3ε on T cells with nanomolar affinity. A serum albumin binding domain was incorporated to extend serum half-life. In this setting, we biophysically characterize and report the activities of CLN-978 in cell co-culture assays, multiple mouse models and non-human primates. RESULTS Human T cells redirected by CLN-978 could eliminate target cells expressing less than 300 copies of CD19 on their surface. The half-life extension and high affinity for CD19 led to significant antitumor activity in murine lymphoma models at very low doses of CLN-978. In primates, we observed a long serum half-life, deep and sustained depletion of normal B cells, and remarkable tolerability, in particular, reduced cytokine release when CLN-978 was administered subcutaneously. CONCLUSIONS CLN-978 warrants further exploration. An ongoing clinical phase 1 trial is investigating safety, pharmacokinetics, pharmacodynamics, and the initial therapeutic potential of subcutaneously administered CLN-978 in patients with non-Hodgkin's lymphoma.
Collapse
Affiliation(s)
| | | | - Bochong Li
- Cullinan Oncology Inc, Cambridge, Massachusetts, USA
| | | | - Patrick A Baeuerle
- Cullinan Oncology Inc, Cambridge, Massachusetts, USA
- Institute of Immunology, Ludwig-Maximilians-Universitat Munchen, Planegg, Germany
| |
Collapse
|
14
|
Isabelle C, Johnson WT, McConnell K, Vogel A, Brammer JE, Boles A, Keller R, Sindaco P, Nisenfeld L, Uppal G, Nikbakht N, Calabretta B, Porazzi P, Gong J, Chakravarti N, Porcu P, Mishra A. Preclinical evaluation of anti-CD38 therapy in mature T-cell neoplasms. Blood Adv 2023; 7:3637-3641. [PMID: 36989058 PMCID: PMC10365939 DOI: 10.1182/bloodadvances.2023009807] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2023] [Revised: 03/09/2023] [Accepted: 03/09/2023] [Indexed: 03/30/2023] Open
Affiliation(s)
- Colleen Isabelle
- Sidney Kimmel Cancer Center, Division of Hematologic Malignancies, Thomas Jefferson University, Philadelphia, PA
| | - William T. Johnson
- Department of Medicine, Lymphoma Service, Memorial Sloan Kettering Cancer Center, New York, NY
| | - Kathleen McConnell
- Sidney Kimmel Cancer Center, Division of Hematologic Malignancies, Thomas Jefferson University, Philadelphia, PA
| | - Ashley Vogel
- Department of Pathology, Anatomy, and Cell Biology, Thomas Jefferson University, Philadelphia, PA
| | - Jonathan E. Brammer
- Division of Hematology, Department of Internal Medicine, The James Comprehensive Center, The Ohio State University, Columbus, OH
| | - Amy Boles
- Sidney Kimmel Cancer Center, Division of Hematologic Malignancies, Thomas Jefferson University, Philadelphia, PA
| | - Robyn Keller
- Sidney Kimmel Cancer Center, Division of Hematologic Malignancies, Thomas Jefferson University, Philadelphia, PA
| | - Paola Sindaco
- Sidney Kimmel Cancer Center, Division of Hematologic Malignancies, Thomas Jefferson University, Philadelphia, PA
| | - Liam Nisenfeld
- Department of Pathology, Anatomy, and Cell Biology, Thomas Jefferson University, Philadelphia, PA
| | - Guldeep Uppal
- Department of Pathology, Anatomy, and Cell Biology, Thomas Jefferson University, Philadelphia, PA
| | - Neda Nikbakht
- Department of Dermatology and Cutaneous Biology, Thomas Jefferson University, Philadelphia, PA
| | - Bruno Calabretta
- Department of Cancer Biology, Sidney Kimmel Cancer Center, Thomas Jefferson University, Philadelphia, PA
| | - Patrizia Porazzi
- Division of Hematology/Oncology, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA
- Center for Cellular Immunotherapies, Division of Hematology/Oncology, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA
| | - Jerald Gong
- Department of Pathology, Anatomy, and Cell Biology, Thomas Jefferson University, Philadelphia, PA
| | - Nitin Chakravarti
- Sidney Kimmel Cancer Center, Division of Hematologic Malignancies, Thomas Jefferson University, Philadelphia, PA
| | - Pierluigi Porcu
- Sidney Kimmel Cancer Center, Division of Hematologic Malignancies, Thomas Jefferson University, Philadelphia, PA
| | - Anjali Mishra
- Sidney Kimmel Cancer Center, Division of Hematologic Malignancies, Thomas Jefferson University, Philadelphia, PA
- Department of Cancer Biology, Sidney Kimmel Cancer Center, Thomas Jefferson University, Philadelphia, PA
| |
Collapse
|
15
|
Lu H, Cheng Z, Hu Y, Tang LV. What Can De Novo Protein Design Bring to the Treatment of Hematological Disorders? BIOLOGY 2023; 12:166. [PMID: 36829445 PMCID: PMC9952452 DOI: 10.3390/biology12020166] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 12/14/2022] [Revised: 01/17/2023] [Accepted: 01/18/2023] [Indexed: 01/22/2023]
Abstract
Protein therapeutics have been widely used to treat hematological disorders. With the advent of de novo protein design, protein therapeutics are not limited to ameliorating natural proteins but also produce novel protein sequences, folds, and functions with shapes and functions customized to bind to the therapeutic targets. De novo protein techniques have been widely used biomedically to design novel diagnostic and therapeutic drugs, novel vaccines, and novel biological materials. In addition, de novo protein design has provided new options for treating hematological disorders. Scientists have designed protein switches called Colocalization-dependent Latching Orthogonal Cage-Key pRoteins (Co-LOCKR) that perform computations on the surface of cells. De novo designed molecules exhibit a better capacity than the currently available tyrosine kinase inhibitors in chronic myeloid leukemia therapy. De novo designed protein neoleukin-2/15 enhances chimeric antigen receptor T-cell activity. This new technique has great biomedical potential, especially in exploring new treatment methods for hematological disorders. This review discusses the development of de novo protein design and its biological applications, with emphasis on the treatment of hematological disorders.
Collapse
Affiliation(s)
| | | | | | - Liang V. Tang
- Department of Hematology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| |
Collapse
|
16
|
Câmara AB, Brandão IA. The Non-Hodgkin Lymphoma Treatment and Side Effects: A Systematic Review and Meta-Analysis. Recent Pat Anticancer Drug Discov 2023; 19:PRA-EPUB-128894. [PMID: 36650656 DOI: 10.2174/1574892818666230117151757] [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: 05/27/2022] [Revised: 10/11/2022] [Accepted: 11/11/2022] [Indexed: 01/19/2023]
Abstract
OBJECTIVE This paper aims to review studies regarding side effects found during Non-Hodgkin Lymphoma treatment, to suggest the drug class most associated with these effects, as well as the most prevalent side effect grade. METHODS This review is registered in PROSPERO (IDCRD42022295774) and followed the PICOS strategy and PRISMA guidelines. The search was carried out in the databases PubMed/MEDLINE, Scientific Electronic Library Online, and DOAJ. Medical Subject Headings Terms were used and quantitative studies with conclusive results regarding side effects during the non-Hodgkin lymphoma treatment were selected. Patent information was obtained from google patents. RESULTS Monoclonal antibodies were the main drug class associated with side effects during NHL therapy. The combination of Rituximab (Rituxan®; patent EP1616572B) and iInotuzumab (Besponsa®; patent EP1504035B3) was associated with a higher incidence of thrombocytopenia (p<0.05), while the combination of Rituximab and Venetoclax (Venclexta®; patent CN107089981A) was associated with a higher incidence of neutropenia (p<0.05) when compared to Bendamustine combinations (Treanda ™; patent US20130253025A1). Meta-analysis revealed a high prevalence of grade 3-4 neutropenia and thrombocytopenia in men. Finally, Americans and Canadians experienced a higher prevalence of these side effects, when compared to others nationalities (p<0.05). CONCLUSION Patents regarding the use of monoclonal antibodies in NHL treatment were published in the last year. Monoclonal antibodies associated with neutropenia (grade 3-4) and thrombocytopenia, especially in North American men treated for NHL, and with an average age of 62 years demonstrated importance in this study.
Collapse
Affiliation(s)
- Alice Barros Câmara
- Department of Biophysics and Pharmacology, Federal University of Rio Grande do Norte
| | - Igor Augusto Brandão
- Bioinformatics Multidisciplinary Environment, Federal University of Rio Grande do Norte
| |
Collapse
|
17
|
Lachance S, Bourguignon A, Boisjoly JA, Bouchard P, Ahmad I, Bambace N, Bernard L, Cohen S, Delisle JS, Fleury I, Kiss T, Mollica L, Roy DC, Sauvageau G, Veilleux O, Zehr J, Chagnon M, Roy J. Impact of Implementing a Bendamustine-Based Conditioning Regimen on Outcomes of Autologous Stem Cell Transplantation in Lymphoma while Novel Cellular Therapies Emerge. Transplant Cell Ther 2023; 29:34.e1-34.e7. [PMID: 36243319 DOI: 10.1016/j.jtct.2022.10.003] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2022] [Revised: 09/28/2022] [Accepted: 10/05/2022] [Indexed: 11/07/2022]
Abstract
With the advent of new cellular and targeted therapies, treatment options for relapsed and refractory (r/R) lymphomas have multiplied, and the optimal approach offering the best outcomes remains a matter of passionate debate. High-dose chemotherapy followed by autologous stem cell transplantation (ASCT) is still considered a treatment option for patients with chemosensitive lymphoma when cure is the expected goal. The myeloablative conditioning regimen preceding the stem cell infusion is considered the effective component of this approach. Carmustine (BCNU)-based preparative regimens, such as BEAM and BEAC, are considered the standard of care and have shown efficacy and low nonrelapse mortality (NRM). Comparative studies between conditioning regimens have failed to identify a better option. After a BCNU drug shortage in Canada followed by a steep increase in price, we elected to substitute BCNU for bendamustine (benda) in the preparative regimen. The purpose of this substitution was to improve response while preserving safety and controlling costs. From May 2015 to May 2018, a total of 131 consecutive lymphoma patients received benda-EAM conditioning. These patients were compared with 96 consecutive patients who received BCNU-based conditioning from January 2012 to May 2015. Apart from conditioning, supportive care measures were the same in the 2 groups. Patients receiving benda were older (55.7 years versus 51.1 years; P = .002). The development of grade ≥3 mucositis was more frequent with benda conditioning (39.5% versus 7.8%; P < .001) leading to a greater requirement for parenteral nutrition (48.9% versus 21.9%; P < .001). A transient creatinine increase >1.5 times the upper limit of normal (15.3% versus 4.2%; P < .008) and intensive care unit admission (6.9% versus 1.1%; P < .029) were more frequent with benda; however, there were no between-group differences in cardiac, pulmonary, or liver toxicity and NRM. With a median follow-up of 48 months for the benda group and 60 months for the BCNU group, benda was associated with significantly better progression-free survival (71% versus 61%; P = .040; hazard ratio [HR], 1.6; 95% confidence interval [CI], 1.0 to 2.7) and overall survival (86% vs 71%; P = .0066; HR, 2.6; 95% CI, 1.3 to 5.4) compared with BCNU-based conditioning regimens. While novel therapies emerge, our study demonstrates that benda-EAM is safe and effective and should be considered a valid alternative to BCNU conditioning to improve outcomes of patients with chemosensitive r/R lymphomas undergoing ASCT.
Collapse
Affiliation(s)
- Sylvie Lachance
- Hôpital Maisonneuve-Rosemont, Montreal, Quebec, Canada; Division of Hematology, Oncology, Hematopoietic Cell Transplant and Cellular Therapy, Hôpital Maisonneuve-Rosemont, Montreal, Quebec, Canada; Hopital Maisonneuve-Rosemont, Division of Hematology, Oncology, Hematopoietic Cell Transplant and Cellular therapy, Université de Montréal, Montreal, Quebec, Canada.
| | - Alex Bourguignon
- Division of Hematology and Oncology, Centre Hospitalier de l'Université de Montréal, Montreal, Quebec, Canada; Hopital Maisonneuve-Rosemont, Division of Hematology, Oncology, Hematopoietic Cell Transplant and Cellular therapy, Université de Montréal, Montreal, Quebec, Canada
| | - Josie-Anne Boisjoly
- Division of Hematology and Oncology, Centre Hospitalier Universitaire de Sherbrooke, Sherbrooke, Quebec, Canada
| | - Philippe Bouchard
- Hôpital Maisonneuve-Rosemont, Montreal, Quebec, Canada; Hopital Maisonneuve-Rosemont, Division of Hematology, Oncology, Hematopoietic Cell Transplant and Cellular therapy, Université de Montréal, Montreal, Quebec, Canada
| | - Imran Ahmad
- Hôpital Maisonneuve-Rosemont, Montreal, Quebec, Canada; Division of Hematology, Oncology, Hematopoietic Cell Transplant and Cellular Therapy, Hôpital Maisonneuve-Rosemont, Montreal, Quebec, Canada; Hopital Maisonneuve-Rosemont, Division of Hematology, Oncology, Hematopoietic Cell Transplant and Cellular therapy, Université de Montréal, Montreal, Quebec, Canada
| | - Nadia Bambace
- Hôpital Maisonneuve-Rosemont, Montreal, Quebec, Canada; Division of Hematology, Oncology, Hematopoietic Cell Transplant and Cellular Therapy, Hôpital Maisonneuve-Rosemont, Montreal, Quebec, Canada; Hopital Maisonneuve-Rosemont, Division of Hematology, Oncology, Hematopoietic Cell Transplant and Cellular therapy, Université de Montréal, Montreal, Quebec, Canada
| | - Léa Bernard
- Hôpital Maisonneuve-Rosemont, Montreal, Quebec, Canada; Division of Hematology, Oncology, Hematopoietic Cell Transplant and Cellular Therapy, Hôpital Maisonneuve-Rosemont, Montreal, Quebec, Canada; Hopital Maisonneuve-Rosemont, Division of Hematology, Oncology, Hematopoietic Cell Transplant and Cellular therapy, Université de Montréal, Montreal, Quebec, Canada
| | - Sandra Cohen
- Hôpital Maisonneuve-Rosemont, Montreal, Quebec, Canada; Division of Hematology, Oncology, Hematopoietic Cell Transplant and Cellular Therapy, Hôpital Maisonneuve-Rosemont, Montreal, Quebec, Canada; Hopital Maisonneuve-Rosemont, Division of Hematology, Oncology, Hematopoietic Cell Transplant and Cellular therapy, Université de Montréal, Montreal, Quebec, Canada
| | - Jean-Sébastien Delisle
- Hôpital Maisonneuve-Rosemont, Montreal, Quebec, Canada; Division of Hematology, Oncology, Hematopoietic Cell Transplant and Cellular Therapy, Hôpital Maisonneuve-Rosemont, Montreal, Quebec, Canada; Hopital Maisonneuve-Rosemont, Division of Hematology, Oncology, Hematopoietic Cell Transplant and Cellular therapy, Université de Montréal, Montreal, Quebec, Canada
| | - Isabelle Fleury
- Hôpital Maisonneuve-Rosemont, Montreal, Quebec, Canada; Division of Hematology, Oncology, Hematopoietic Cell Transplant and Cellular Therapy, Hôpital Maisonneuve-Rosemont, Montreal, Quebec, Canada; Hopital Maisonneuve-Rosemont, Division of Hematology, Oncology, Hematopoietic Cell Transplant and Cellular therapy, Université de Montréal, Montreal, Quebec, Canada
| | - Thomas Kiss
- Hôpital Maisonneuve-Rosemont, Montreal, Quebec, Canada; Division of Hematology, Oncology, Hematopoietic Cell Transplant and Cellular Therapy, Hôpital Maisonneuve-Rosemont, Montreal, Quebec, Canada; Hopital Maisonneuve-Rosemont, Division of Hematology, Oncology, Hematopoietic Cell Transplant and Cellular therapy, Université de Montréal, Montreal, Quebec, Canada
| | - Luigina Mollica
- Hôpital Maisonneuve-Rosemont, Montreal, Quebec, Canada; Division of Hematology, Oncology, Hematopoietic Cell Transplant and Cellular Therapy, Hôpital Maisonneuve-Rosemont, Montreal, Quebec, Canada; Hopital Maisonneuve-Rosemont, Division of Hematology, Oncology, Hematopoietic Cell Transplant and Cellular therapy, Université de Montréal, Montreal, Quebec, Canada
| | - Denis-Claude Roy
- Hôpital Maisonneuve-Rosemont, Montreal, Quebec, Canada; Division of Hematology, Oncology, Hematopoietic Cell Transplant and Cellular Therapy, Hôpital Maisonneuve-Rosemont, Montreal, Quebec, Canada; Hopital Maisonneuve-Rosemont, Division of Hematology, Oncology, Hematopoietic Cell Transplant and Cellular therapy, Université de Montréal, Montreal, Quebec, Canada
| | - Guy Sauvageau
- Hôpital Maisonneuve-Rosemont, Montreal, Quebec, Canada; Division of Hematology, Oncology, Hematopoietic Cell Transplant and Cellular Therapy, Hôpital Maisonneuve-Rosemont, Montreal, Quebec, Canada; Hopital Maisonneuve-Rosemont, Division of Hematology, Oncology, Hematopoietic Cell Transplant and Cellular therapy, Université de Montréal, Montreal, Quebec, Canada
| | - Olivier Veilleux
- Hôpital Maisonneuve-Rosemont, Montreal, Quebec, Canada; Division of Hematology, Oncology, Hematopoietic Cell Transplant and Cellular Therapy, Hôpital Maisonneuve-Rosemont, Montreal, Quebec, Canada; Hopital Maisonneuve-Rosemont, Division of Hematology, Oncology, Hematopoietic Cell Transplant and Cellular therapy, Université de Montréal, Montreal, Quebec, Canada
| | - Justine Zehr
- Department of Medicine and Biostatistics, Université de Montréal, Montreal, Quebec, Canada; Hopital Maisonneuve-Rosemont, Division of Hematology, Oncology, Hematopoietic Cell Transplant and Cellular therapy, Université de Montréal, Montreal, Quebec, Canada
| | - Miguel Chagnon
- Department of Medicine and Biostatistics, Université de Montréal, Montreal, Quebec, Canada; Hopital Maisonneuve-Rosemont, Division of Hematology, Oncology, Hematopoietic Cell Transplant and Cellular therapy, Université de Montréal, Montreal, Quebec, Canada
| | - Jean Roy
- Hôpital Maisonneuve-Rosemont, Montreal, Quebec, Canada; Division of Hematology, Oncology, Hematopoietic Cell Transplant and Cellular Therapy, Hôpital Maisonneuve-Rosemont, Montreal, Quebec, Canada; Hopital Maisonneuve-Rosemont, Division of Hematology, Oncology, Hematopoietic Cell Transplant and Cellular therapy, Université de Montréal, Montreal, Quebec, Canada
| |
Collapse
|
18
|
Nogami A, Sasaki K. Therapeutic Advances in Immunotherapies for Hematological Malignancies. Int J Mol Sci 2022; 23:11526. [PMID: 36232824 PMCID: PMC9569660 DOI: 10.3390/ijms231911526] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2022] [Revised: 09/21/2022] [Accepted: 09/26/2022] [Indexed: 11/23/2022] Open
Abstract
Following the success of immunotherapies such as chimeric antigen receptor transgenic T-cell (CAR-T) therapy, bispecific T-cell engager therapy, and immune checkpoint inhibitors in the treatment of hematologic malignancies, further studies are underway to improve the efficacy of these immunotherapies and to reduce the complications associated with their use in combination with other immune checkpoint inhibitors and conventional chemotherapy. Studies of novel therapeutic strategies such as bispecific (tandem or dual) CAR-T, bispecific killer cell engager, trispecific killer cell engager, and dual affinity retargeting therapies are also underway. Because of these studies and the discovery of novel immunotherapeutic target molecules, the use of immunotherapy for diseases initially thought to be less promising to treat with this treatment method, such as acute myeloid leukemia and T-cell hematologic tumors, has become a reality. Thus, in this coming era of new transplantation- and chemotherapy-free treatment strategies, it is imperative for both scientists and clinicians to understand the molecular immunity of hematologic malignancies. In this review, we focus on the remarkable development of immunotherapies that could change the prognosis of hematologic diseases. We also review the molecular mechanisms, development processes, clinical efficacies, and problems of new agents.
Collapse
Affiliation(s)
- Ayako Nogami
- Department of Laboratory Medicine, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University, 1-5-45 Yushima, Bunkyoku, Tokyo 1138510, Japan
- Department of Hematology, Tokyo Medical and Dental University Hospital, 1-5-45 Yushima, Bunkyoku, Tokyo 1138510, Japan
| | - Koji Sasaki
- Department of Leukemia, The University of Texas MD Anderson Cancer Center, 1515 Holcombe Boulevard, Unit 428, Houston, TX 77030, USA
| |
Collapse
|
19
|
Abstract
PURPOSE OF REVIEW T-cell-engaging antibodies or T-cell engagers (TCEs) can connect a patient's cytotoxic T cells with cancer cells, leading to potent redirected lysis. Until very recently, only one TCE was approved, the CD19/CD3-bispecific blinatumomab. Many new TCEs in late-stage clinical development target various hematopoietic lineage markers like CD20, BCMA, or CD123. Although very compelling single-agent activity of TCEs was observed with various blood-borne cancers, therapy of solid tumor indications has thus far been less successful. RECENT FINDINGS The approval in 2022 of the gp100 peptide-major histocompatibility complex (MHC)/CD3 bispecific TCE tebentafusp in uveal melanoma confirms that TCEs can also efficiently work against solid tumors. TCEs targeting peptide-MHC complexes will expand the target space for solid tumor therapy to intracellular targets. Likewise, early clinical trial data from TCEs targeting DLL3 in small cell lunger cancer showed promising antitumor activity. Various technologies for conditional activation of TCEs in the tumor microenvironment (TME) may expand the scope of conventional surface targets that suffer from a narrow therapeutic window. Finally, pharmacological enhancements for TCE therapies by engagement of certain costimulatory receptors and cytokines, or blockade of checkpoints, are showing promise. SUMMARY Targeting peptide-MHC complexes, conditional TCE technologies, and concepts enhancing TCE-activated T cells are paving the way towards overcoming challenges associated with solid tumor therapy.
Collapse
|
20
|
Ibrutinib Associated with Rituximab-Platinum Salt-Based Immunochemotherapy in B-Cell Lymphomas: Results of a Phase 1b-II Study of the LYSA Group. Cancers (Basel) 2022; 14:cancers14071761. [PMID: 35406532 PMCID: PMC8997053 DOI: 10.3390/cancers14071761] [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: 02/13/2022] [Revised: 03/18/2022] [Accepted: 03/25/2022] [Indexed: 11/29/2022] Open
Abstract
Simple Summary Patients with relapsing/refractory B-cell lymphoma who respond to a platinum salt-based salvage regimen can be cured after therapy intensification followed by autologous stem cell transplantation. The Bruton tyrosine kinase inhibitor ibrutinib, given alone or in association with other molecules, has proven effective in numerous B-cell lymphomas. The aim of the current study was to evaluate the safety of the combination of ibrutinib, rituximab, dexamethasone, and cytarabine with either cisplatin (R-DHAP) or oxaliplatin (R-DHAOx), with ibrutinib given based on a prespecified dose-escalation design. The concomitant combination of ibrutinib and R-DHAP resulted in limiting hematological, infectious, and renal toxicities. As a result, the maximum ibrutinib dose could not be administered. On the other hand, when the ibrutinib administration schedule was combined with R-DHAOx, ibrutinib dosing could be increased up to the maximum prespecified dose but with relevant toxicities. Despite a strong rationale for combining ibrutinib with either R-DHAP or R-DHAP/Ox, this approach was limited by significant toxicities. Abstract In the post-rituximab era, patients with relapsed/refractory non-Hodgkin B-cell lymphoma (R/R B-NHL) responding to a platinum salt-based salvage regimen can potentially be cured after intensification followed by autologous stem cell transplantation, with the quality of the response to salvage predicting survival. The Bruton tyrosine kinase inhibitor ibrutinib, given as monotherapy or combined with other molecules, has proven effective in numerous B-cell lymphomas. To evaluate the safety of the combination of ibrutinib, rituximab, dexamethasone, and cytarabine with either cisplatin (R-DHAP) or oxaliplatin (R-DHAOx), we conducted a multicenter Phase 1b-II study in transplant-eligible R/R B-NHL patients, with ibrutinib given using a 3-by-3 dose-escalation design. The combination of R-DHAP and ibrutinib (given from Day 1 to Day 21 of each cycle) was associated with dose-limiting hematological, infectious, and renal toxicities, while we were unable to reach a dose to recommend for Phase II. R-DHAOx could only be combined with a daily dosage of 280 mg ibrutinib when administered continuously. R-DHAP combined with intermittent ibrutinib administration (from Day 5 to Day 18) was found to be highly toxic. On the other hand, when this administration schedule was combined with R-DHAOx, ibrutinib dosing could be increased up to 560 mg but with relevant toxicities. Despite a strong rationale for combining ibrutinib and R-DHAP/R-DHAOx, as both target lymphoma B-cells by different mechanisms, this approach was limited by significant toxicities.
Collapse
|
21
|
Rai S, Tanizawa Y, Cai Z, Huang YJ, Taipale K, Tajimi M. Outcomes for Recurrent Mantle Cell Lymphoma Post-Ibrutinib Therapy: A Retrospective Cohort Study from a Japanese Administrative Database. Adv Ther 2022; 39:4792-4807. [PMID: 35984628 PMCID: PMC9464745 DOI: 10.1007/s12325-022-02258-3] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2022] [Accepted: 07/01/2022] [Indexed: 01/30/2023]
Abstract
INTRODUCTION Treatment options in patients with mantle cell lymphoma (MCL) failing ibrutinib are limited, with no standard therapies defined. This study aimed to investigate real-world treatment patterns and outcomes for patients with MCL following ibrutinib. METHODS This study utilized a de-identified hospital-based claims database (Medical Data Vision) in Japan. Eligible patients were adults who were diagnosed with MCL and had received antitumor drugs between December 2010 and July 2020. Patients were followed from the first antitumor drug treatment until the end of available data up to July 2021. Time-to-event analyses utilized the Kaplan-Meier method. Factors for receiving post-ibrutinib therapy were explored with logistic regression analysis. RESULTS Of the 1386 patients who started antitumor drug therapy, 247 patients received and discontinued ibrutinib at any line of therapy. Among them, 137 patients (55.5%) received subsequent therapy. The median age at the end of ibrutinib therapy was 77 (range 42-95), and 44 patients had a dependent activity of daily living (ADL). Factors negatively associated with receiving post-ibrutinib therapy after discontinuation of ibrutinib were age ≥ 75 years (odds ratio [95% CI] 0.46 [0.26-0.80]) and emergency hospital admissions (0.37 [0.17-0.84]). Immediate post-ibrutinib therapy regimens were highly diverse, with BR (bendamustine, rituximab) only prescribed in more than 10% of patients. The median duration of post-ibrutinib therapy was 1.5 months (95% CI 1.07-2.07). The median overall survival from the end of ibrutinib therapy in patients regardless of the receipt of post-ibrutinib therapy (n = 247), in those who did not receive post-ibrutinib therapy (n = 110), and in those who received post-ibrutinib therapy (n = 137) was 5.6 months (95% CI 3.8-8.7), 2.3 months (95% CI 1.2-3.9), and 8.7 months (95% CI 5.6-13.8), respectively. The most common adverse event during post-ibrutinib therapy was infection, with the use of anti-infectives (17%). CONCLUSIONS Patients with MCL previously treated with ibrutinib have poor ability to carry out ADL and experience very poor outcomes. New safe, effective therapies are needed.
Collapse
Affiliation(s)
- Shinya Rai
- Department of Hematology and Rheumatology, Faculty of Medicine, Kindai University, Osaka, Sayama, Japan.
| | | | | | | | | | | |
Collapse
|