1
|
Long AH, Aftandilian C, Barmettler S, Alexander S. Hypogammaglobulinemia in Children Receiving Targeted Immunotherapies for B Lineage Malignancies: Practical Guidance for Assessment and Management. Pediatr Blood Cancer 2025:e31779. [PMID: 40372257 DOI: 10.1002/pbc.31779] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/11/2025] [Revised: 04/21/2025] [Accepted: 04/24/2025] [Indexed: 05/16/2025]
Abstract
Hypogammaglobulinemia is a well-defined risk factor for infection. B-cell-directed immunotherapies given in addition to conventional chemotherapy are now core elements of effective therapy for children with B lymphoid malignancies. These therapies are associated with depletion of normal B cells and consequent hypogammaglobulinemia. This review summarizes the current state of knowledge regarding the mechanism, incidence, and clinical outcomes related to hypogammaglobulinemia in children with mature B-cell non-Hodgkin lymphoma and B-cell acute lymphoblastic leukemia, as well as provides practical guidance for laboratory monitoring and considerations for immunoglobulin replacement therapy.
Collapse
Affiliation(s)
- Adrienne H Long
- Division of Hematology, Oncology, Stem Cell Transplantation, and Regenerative Medicine, Department of Pediatrics, Stanford University, Stanford, California, USA
| | - Catherine Aftandilian
- Division of Hematology, Oncology, Stem Cell Transplantation, and Regenerative Medicine, Department of Pediatrics, Stanford University, Stanford, California, USA
| | - Sara Barmettler
- Division of Rheumatology, Allergy and Immunology, Department of Medicine, Massachusetts General Hospital, Boston, Massachusetts, USA
| | - Sarah Alexander
- Division of Haematology/Oncology, The Hospital for Sick Children, Toronto, Ontario, Canada
| |
Collapse
|
2
|
Kacar M, Al-Hakim A, Savic S. Sequelae of B-Cell Depleting Therapy: An Immunologist's Perspective. BioDrugs 2025; 39:103-130. [PMID: 39680306 DOI: 10.1007/s40259-024-00696-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 11/27/2024] [Indexed: 12/17/2024]
Abstract
B-cell depleting therapy (BCDT) has revolutionised the treatment of B-cell malignancies and autoimmune diseases by targeting specific B-cell surface antigens, receptors, ligands, and signalling pathways. This narrative review explores the mechanisms, applications, and complications of BCDT, focusing on the therapeutic advancements since the introduction of rituximab in 1997. Various monoclonal antibodies and kinase inhibitors are examined for their roles in depleting B cells through antibody-dependent and independent mechanisms. The off-target effects, such as hypogammaglobulinemia, infections, and cytokine release syndrome, are discussed, emphasising the need for immunologists to identify and help manage these complications. The increasing prevalence of BCDT has necessitated the involvement of clinical immunologists in addressing treatment-associated immunological abnormalities, including persistent hypogammaglobulinemia and neutropenia. We highlight the importance of considering underlying inborn errors of immunity (IEI) in patients presenting with these complications. Furthermore, we discuss the impact of BCDT on other immune cell populations and the challenges in predicting and managing long-term immunological sequelae. The potential for novel BCDT agents targeting the BAFF/APRIL-TACI/BCMA axis and B-cell receptor signalling pathways to treat autoimmune disorders is also explored, underscoring the rapidly evolving landscape of B-cell targeted therapies.
Collapse
Affiliation(s)
- Mark Kacar
- Department of Allergy, University Clinic Golnik, Golnik, Slovenia
- Department of Allergy and Clinical Immunology, St James' University Hospital, Leeds, UK
- Faculty of Medicine, University of Ljubljana, Ljubljana, Slovenia
| | - Adam Al-Hakim
- Department of Allergy and Clinical Immunology, St James' University Hospital, Leeds, UK
- Leeds Institute of Rheumatic and Musculoskeletal Medicine, University of Leeds, Leeds, UK
| | - Sinisa Savic
- Department of Allergy and Clinical Immunology, St James' University Hospital, Leeds, UK.
- Leeds Institute of Rheumatic and Musculoskeletal Medicine, University of Leeds, Leeds, UK.
- NIHR Leeds Biomedical Research Centre, Leeds, UK.
| |
Collapse
|
3
|
Elmoursi A, Barmettler S. Therapeutic B-cell depletion: Mechanisms, clinical applications, and implications for secondary immunodeficiency. J Allergy Clin Immunol 2024:S0091-6749(24)01278-8. [PMID: 39613109 DOI: 10.1016/j.jaci.2024.11.026] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2024] [Revised: 11/16/2024] [Accepted: 11/21/2024] [Indexed: 12/01/2024]
Affiliation(s)
- Ahmed Elmoursi
- Division of Rheumatology, Allergy, and Immunology, Department of Medicine, Massachusetts General Hospital, Boston, Mass; Harvard Medical School, Boston, Mass
| | - Sara Barmettler
- Division of Rheumatology, Allergy, and Immunology, Department of Medicine, Massachusetts General Hospital, Boston, Mass; Harvard Medical School, Boston, Mass.
| |
Collapse
|
4
|
Sayyed A, Wilson L, Stavi V, Chen S, Chen C, Mattsson J, Lipton JH, Kim DD, Viswabandya A, Kumar R, Lam W, Law AD, Gerbitz A, Pasic I, Novitzky-Basso I, Mazzulli T, Michelis FV. Impact of cytomegalovirus (CMV) seroconversion pre-allogeneic hematopoietic cell transplantation on posttransplant outcomes. Eur J Haematol 2024; 113:441-453. [PMID: 38880946 DOI: 10.1111/ejh.14251] [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: 03/14/2024] [Revised: 05/22/2024] [Accepted: 05/23/2024] [Indexed: 06/18/2024]
Abstract
Cytomegalovirus (CMV) reactivation post-allogeneic hematopoietic cell transplantation (post-alloHCT) increases morbidity and mortality. We sought to determine the frequency of CMV seroconversion in patients pre-alloHCT and to investigate the impact on posttransplant outcomes. We retrospectively investigated 752 adult patients who underwent alloHCT at our center from January 2015 to February 2020 before the adoption of letermovir prophylaxis. CMV serology was assessed at consult and pretransplant. The cohort was divided into four groups based on pretransplant CMV seroconversion: negative to positive (Group 1), positive to negative (Group 2), consistently negative (Group 3), and consistently positive (Group 4). Eighty-nine patients (12%) had seroconverted from negative to positive, 17 (2%) from positive to negative, 151 (20%) were consistently seronegative, and 495 (66%) were consistently seropositive pretransplant. For the four CMV serostatus groups, cumulative incidence of CMV reactivation at 6 months posttransplant was 4.5%, 47.1%, 6.6%, and 76.6% for Groups 1, 2, 3, and 4, respectively (p < .0001). No differences between groups were seen regarding Grade III-IV acute graft-versus-host disease (GVHD) (p = .91), moderate/severe chronic GVHD (p = .41), or graft failure (p = .28). On multivariable analysis, there was no impact of CMV serostatus group on overall survival (p = .67), cumulative incidence of relapse (p = .83) or non-relapse mortality. alloHCT patients who demonstrate CMV seroconversion pretransplant from negative to positive have a very low risk of CMV reactivation posttransplant. The observed seroconversion may be due to passive CMV immunity acquired through blood products. Quantitative CMV immunoglobulin G/immunoglobulin M pretransplant may help differentiate between true seroconversion and passively transmitted CMV immunoglobulin.
Collapse
Affiliation(s)
- Ayman Sayyed
- Hans Messner Allogeneic Transplant Program, Division of Medical Oncology and Hematology, Princess Margaret Cancer Centre, University Health Network, Toronto, Ontario, Canada
| | - Leeann Wilson
- Hans Messner Allogeneic Transplant Program, Division of Medical Oncology and Hematology, Princess Margaret Cancer Centre, University Health Network, Toronto, Ontario, Canada
| | - Vered Stavi
- Hans Messner Allogeneic Transplant Program, Division of Medical Oncology and Hematology, Princess Margaret Cancer Centre, University Health Network, Toronto, Ontario, Canada
| | - Shiyi Chen
- Department of Biostatistics, Princess Margaret Cancer Centre, University Health Network, Toronto, Canada
| | - Carol Chen
- Hans Messner Allogeneic Transplant Program, Division of Medical Oncology and Hematology, Princess Margaret Cancer Centre, University Health Network, Toronto, Ontario, Canada
| | - Jonas Mattsson
- Hans Messner Allogeneic Transplant Program, Division of Medical Oncology and Hematology, Princess Margaret Cancer Centre, University Health Network, Toronto, Ontario, Canada
| | - Jeffrey H Lipton
- Hans Messner Allogeneic Transplant Program, Division of Medical Oncology and Hematology, Princess Margaret Cancer Centre, University Health Network, Toronto, Ontario, Canada
| | - Dennis D Kim
- Hans Messner Allogeneic Transplant Program, Division of Medical Oncology and Hematology, Princess Margaret Cancer Centre, University Health Network, Toronto, Ontario, Canada
| | - Auro Viswabandya
- Hans Messner Allogeneic Transplant Program, Division of Medical Oncology and Hematology, Princess Margaret Cancer Centre, University Health Network, Toronto, Ontario, Canada
| | - Rajat Kumar
- Hans Messner Allogeneic Transplant Program, Division of Medical Oncology and Hematology, Princess Margaret Cancer Centre, University Health Network, Toronto, Ontario, Canada
| | - Wilson Lam
- Hans Messner Allogeneic Transplant Program, Division of Medical Oncology and Hematology, Princess Margaret Cancer Centre, University Health Network, Toronto, Ontario, Canada
| | - Arjun D Law
- Hans Messner Allogeneic Transplant Program, Division of Medical Oncology and Hematology, Princess Margaret Cancer Centre, University Health Network, Toronto, Ontario, Canada
| | - Armin Gerbitz
- Hans Messner Allogeneic Transplant Program, Division of Medical Oncology and Hematology, Princess Margaret Cancer Centre, University Health Network, Toronto, Ontario, Canada
| | - Ivan Pasic
- Hans Messner Allogeneic Transplant Program, Division of Medical Oncology and Hematology, Princess Margaret Cancer Centre, University Health Network, Toronto, Ontario, Canada
| | - Igor Novitzky-Basso
- Hans Messner Allogeneic Transplant Program, Division of Medical Oncology and Hematology, Princess Margaret Cancer Centre, University Health Network, Toronto, Ontario, Canada
| | - Tony Mazzulli
- Department of Microbiology, Sinai Health System/University Health Network, Toronto, Ontario, Canada
| | - Fotios V Michelis
- Hans Messner Allogeneic Transplant Program, Division of Medical Oncology and Hematology, Princess Margaret Cancer Centre, University Health Network, Toronto, Ontario, Canada
| |
Collapse
|
5
|
Wang YL, Chang TY, Wen YC, Yang SH, Hsiao YW, Chiu CC, Chen YC, Hu RS, Chen SH, Jaing TH, Hsiao CC. Blinatumomab in Children with MRD-Positive B-Cell Precursor Acute Lymphoblastic Leukemia: A Report of 11 Cases. Hematol Rep 2024; 16:347-353. [PMID: 38921183 PMCID: PMC11204057 DOI: 10.3390/hematolrep16020035] [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: 08/09/2023] [Revised: 05/01/2024] [Accepted: 05/28/2024] [Indexed: 06/27/2024] Open
Abstract
Background/Objectives: Relapsed B-cell acute lymphoblastic leukemia (B-ALL) remains an unresolved matter of concern regarding adverse outcomes. This case study aimed to evaluate the effectiveness of blinatumomab, with or without door lymphocyte infusion (DLI), in treating measurable residual disease (MRD)-positive B-ALL. Methods: All patients who received blinatumomab salvage therapy were included in this study. Eleven patients were included in the study. All patients were evaluated for MRD-negativity. Results: Before starting blinatumomab therapy, seven patients tested positive for MRD, three tested negative, and one had refractory disease. Hematopoietic cell transplantation (HCT) was reserved for five patients with persistent MRD. Six patients became MRD-negative and subsequent HCT was not performed. Only two patients relapsed; one patient died of relapse, and the other one received carfilzomib-based therapy and was MRD-negative thereafter. Nine patients were MRD-negative at a median follow-up of 28 months (15-52 months). Two of three MRD-positive post-transplant patients remained in complete molecular remission after preemptive DLI at the last follow-up date. In the first salvage, blinatumomab may achieve complete remission and bridging to HCT in pediatric patients with end-of-induction MRD-positive B-cell precursor ALL. Conclusions: The decision on how to treat post-transplant relapse continues to affect survival outcomes. Blinatumomab combined with DLI may extend the armamentarium of release options for high-risk pediatric patients. This approach is encouraging for high-risk ALL patients who are MRD-positive post-transplantation.
Collapse
Affiliation(s)
- Yi-Lun Wang
- Department of Pediatrics, Division of Hematology/Oncology, Chang Gung Memorial Hospital, Taoyuan 33315, Taiwan; (Y.-L.W.); (T.-Y.C.); (S.-H.C.)
| | - Tsung-Yen Chang
- Department of Pediatrics, Division of Hematology/Oncology, Chang Gung Memorial Hospital, Taoyuan 33315, Taiwan; (Y.-L.W.); (T.-Y.C.); (S.-H.C.)
| | - Yu-Chuan Wen
- Department of Nursing, Chang Gung Memorial Hospital, Taoyuan 33315, Taiwan; (Y.-C.W.); (S.-H.Y.); (Y.-W.H.); (C.-C.C.)
| | - Shu-Ho Yang
- Department of Nursing, Chang Gung Memorial Hospital, Taoyuan 33315, Taiwan; (Y.-C.W.); (S.-H.Y.); (Y.-W.H.); (C.-C.C.)
| | - Yi-Wen Hsiao
- Department of Nursing, Chang Gung Memorial Hospital, Taoyuan 33315, Taiwan; (Y.-C.W.); (S.-H.Y.); (Y.-W.H.); (C.-C.C.)
| | - Chia-Chi Chiu
- Department of Nursing, Chang Gung Memorial Hospital, Taoyuan 33315, Taiwan; (Y.-C.W.); (S.-H.Y.); (Y.-W.H.); (C.-C.C.)
| | - Yu-Chieh Chen
- Department of Pediatrics, Kaohsiung Chang Gung Memorial Hospital, Kaohsiung 83301, Taiwan; (Y.-C.C.); (C.-C.H.)
- College of Medicine, Chang Gung University, Taoyuan 33302, Taiwan;
| | - Ruei-Shan Hu
- College of Medicine, Chang Gung University, Taoyuan 33302, Taiwan;
| | - Shih-Hsiang Chen
- Department of Pediatrics, Division of Hematology/Oncology, Chang Gung Memorial Hospital, Taoyuan 33315, Taiwan; (Y.-L.W.); (T.-Y.C.); (S.-H.C.)
- College of Medicine, Chang Gung University, Taoyuan 33302, Taiwan;
| | - Tang-Her Jaing
- Department of Pediatrics, Division of Hematology/Oncology, Chang Gung Memorial Hospital, Taoyuan 33315, Taiwan; (Y.-L.W.); (T.-Y.C.); (S.-H.C.)
- College of Medicine, Chang Gung University, Taoyuan 33302, Taiwan;
| | - Chih-Cheng Hsiao
- Department of Pediatrics, Kaohsiung Chang Gung Memorial Hospital, Kaohsiung 83301, Taiwan; (Y.-C.C.); (C.-C.H.)
- College of Medicine, Chang Gung University, Taoyuan 33302, Taiwan;
| |
Collapse
|
6
|
Subklewe M, Magno G, Gebhardt C, Bücklein V, Szelinski F, Arévalo HJR, Hänel G, Dörner T, Zugmaier G, von Bergwelt-Baildon M, Skapenko A, Schulze-Koops H. Application of blinatumomab, a bispecific anti-CD3/CD19 T-cell engager, in treating severe systemic sclerosis: A case study. Eur J Cancer 2024; 204:114071. [PMID: 38691878 DOI: 10.1016/j.ejca.2024.114071] [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: 04/18/2024] [Accepted: 04/18/2024] [Indexed: 05/03/2024]
Abstract
Systemic sclerosis, a severe inflammatory autoimmune disease, shares a common thread with cancer through the underlying mechanism of inflammation. This inflammatory milieu not only drives the immune dysregulation characteristic of autoimmune diseases but also plays a pivotal role in the pathogenesis of cancer. Among the cellular components involved, B cells have emerged as key players in hematologic tumor and autoimmune disease, contributing to immune dysregulation and persistent tissue fibrosis in systemic sclerosis, as well as tumor progression and immune evasion in cancer. Consequently, novel therapeutic strategies targeting B cells hold promise in both conditions. Recent exploration of CD19 CAR T cells in severe systemic sclerosis patients has shown great potential, but also introduced possible risks and drawbacks associated with viral vectors, prolonged CAR T cell persistence, lengthy production timelines, high costs, and the necessity of conditioning patients with organotoxic and fertility-damaging chemotherapy. Given these challenges, alternative CD19-depleting approaches are of high interest for managing severe systemic autoimmune diseases. Here, we present the pioneering use of blinatumomab, a bispecific anti-CD3/anti-CD19 T cell engager in a patient with progressive, severe systemic sclerosis, offering a promising alternative for such challenging cases.
Collapse
Affiliation(s)
- Marion Subklewe
- Department of Medicine III - Hematology/Oncology, LMU University Hospital, LMU Munich, Munich, Germany; Laboratory for Translational Cancer Immunology, LMU Gene Center, Munich, Germany; German Cancer Consortium (DKTK), Munich and Berlin sites, and German Cancer Research Center, Heidelberg, Germany.
| | - Giulia Magno
- Department of Medicine III - Hematology/Oncology, LMU University Hospital, LMU Munich, Munich, Germany.
| | - Christina Gebhardt
- Division of Rheumatology and Clinical Immunology, Department of Medicine IV, LMU University Hospital, LMU Munich, Munich, Germany.
| | - Veit Bücklein
- Department of Medicine III - Hematology/Oncology, LMU University Hospital, LMU Munich, Munich, Germany; Laboratory for Translational Cancer Immunology, LMU Gene Center, Munich, Germany; German Cancer Consortium (DKTK), Munich and Berlin sites, and German Cancer Research Center, Heidelberg, Germany.
| | - Franziska Szelinski
- Department of Medicine, Rheumatology and Clinical Immunology, Charite University Hospital, Berlin, Germany.
| | | | - Gerulf Hänel
- Laboratory for Translational Cancer Immunology, LMU Gene Center, Munich, Germany.
| | - Thomas Dörner
- Department of Medicine, Rheumatology and Clinical Immunology, Charite University Hospital, Berlin, Germany.
| | | | - Michael von Bergwelt-Baildon
- Department of Medicine III - Hematology/Oncology, LMU University Hospital, LMU Munich, Munich, Germany; German Cancer Consortium (DKTK), Munich and Berlin sites, and German Cancer Research Center, Heidelberg, Germany.
| | - Alla Skapenko
- Division of Rheumatology and Clinical Immunology, Department of Medicine IV, LMU University Hospital, LMU Munich, Munich, Germany.
| | - Hendrik Schulze-Koops
- Division of Rheumatology and Clinical Immunology, Department of Medicine IV, LMU University Hospital, LMU Munich, Munich, Germany.
| |
Collapse
|
7
|
Brivio E, Bautista F, Zwaan CM. Naked antibodies and antibody-drug conjugates: targeted therapy for childhood acute lymphoblastic leukemia. Haematologica 2024; 109:1700-1712. [PMID: 38832425 PMCID: PMC11141655 DOI: 10.3324/haematol.2023.283815] [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: 03/15/2024] [Indexed: 06/05/2024] Open
Abstract
The treatment of childhood acute lymphoblastic leukemia (ALL) has reached overall survival rates exceeding 90%. The present and future challenges are to cure the remainder of patients still dying from disease, and to reduce morbidity and mortality in those who can be cured with standard-of-care chemotherapy by replacing toxic chemotherapy elements while retaining cure rates. With the novel therapeutic options introduced in the last years, including immunotherapies and targeted antibodies, the treatment of ALL is undergoing major changes. For B-cell precursor ALL, blinatumomab, an anti-CD19 bispecific antibody, has established its role in the consolidation treatment for both high- and standard-risk first relapse of ALL, in the presence of bone marrow involvement, and may also have an impact on the outcome of high-risk subsets such as infant ALL and Philadelphia chromosome-positive ALL. Inotuzumab ozogamicin, an anti-CD22 drug conjugated antibody, has demonstrated high efficacy in inducing complete remission in relapsed ALL, even in the presence of high tumor burden, but randomized phase III trials are still ongoing. For T-ALL the role of CD38-directed treatment, such as daratumumab, is gaining interest, but randomized data are needed to assess its specific benefit. These antibodies are currently being tested in patients with newly diagnosed ALL and may lead to major changes in the present paradigm of treatment of pediatric ALL. Unlike the past, lessons may be learned from innovations in adult ALL, in which more drastic changes are piloted that may need to be translated to pediatrics.
Collapse
Affiliation(s)
- Erica Brivio
- Princess Máxima Center for Pediatric Oncology, Utrecht
| | | | - C. Michel Zwaan
- Princess Máxima Center for Pediatric Oncology, Utrecht
- Pediatric Oncology, Erasmus MC-Sophia Children’s Hospital, Rotterdam, the Netherlands
| |
Collapse
|
8
|
Shah M, El Chaer F, Ho DY, El Boghdadly Z. Managing infectious challenges in the age of molecular-targeted therapies for adult hematological malignancies. Transpl Infect Dis 2024; 26:e14283. [PMID: 38698640 DOI: 10.1111/tid.14283] [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: 11/15/2023] [Revised: 03/15/2024] [Accepted: 04/02/2024] [Indexed: 05/05/2024]
Abstract
Over the last decade, the therapeutic landscape for hematological malignancies (HMs) has witnessed a remarkable surge in the development of novel biological and small-molecule-targeted immunomodulatory agents. These therapies have drastically improved survival, but some come at the cost of increased risk of bacterial, viral, and/or fungal infections and on-target off-tumor immunological side effects. To mitigate such risks, physicians must be well informed about infectious complications and necessary preventive measures, such as screening, vaccinations, and antimicrobial prophylaxis. Furthermore, physicians should be vigilant about the noninfectious side effects of these agents that can mimic infections and understand their potential drug-drug interactions with antimicrobials. Strengthening and harmonizing the current surveillance and reporting system for drug-associated infections in real-world settings is essential to better ascertain the potential infections associated with these agents. In this review, we aimed to summarize the infection risks associated with novel agents used for specific HMs and outline recommended strategies for monitoring and prophylaxis.
Collapse
Affiliation(s)
- Manan Shah
- Division of Hematology and Medical Oncology, Department of Internal Medicine, The Ohio State University, Columbus, Ohio, USA
| | - Firas El Chaer
- Division of Hematology and Oncology, Department of Medicine, The University of Virginia, Charlottesville, Virginia, USA
| | - Dora Y Ho
- Division of Infectious Diseases and Geographic Medicine, Department of Medicine, Stanford University, Stanford, Virginia, USA
| | - Zeinab El Boghdadly
- Division of Infectious Diseases, Department of Internal Medicine, The Ohio State University, Columbus, Ohio, USA
| |
Collapse
|
9
|
Lantz J, Pham N, Jones C, Reed D, El Chaer F, Keng M. Blinatumomab in Practice. Curr Hematol Malig Rep 2024; 19:1-8. [PMID: 38060085 DOI: 10.1007/s11899-023-00714-7] [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: 10/02/2023] [Indexed: 12/08/2023]
Abstract
PURPOSE OF REVIEW Acute lymphoblastic leukemia (ALL) is a rare hematologic neoplasm in adults, with most cases defined by pathology related to abnormal B cell proliferation known as B-cell ALL. The course is challenging, with less-than-optimal survival outcomes, even with aggressive multiagent chemotherapy and consideration for stem cell transplantation. Novel therapies focused on targetable pathways are being investigated to improve outcomes while simultaneously decreasing toxicity. In our review, we aim to evaluate the utilization of blinatumomab in B-cell ALL and provide insight on how this guides our management. RECENT FINDINGS Blinatumomab is a bispecific T-cell engager (BiTE) immunotherapy that neutralizes malignant cells by instigating CD3-positive T cells to target CD19-positive B cells. However, this therapy targets both malignant and non-malignant lymphocytes with potentially severe side effects such as cytokine release syndrome or neurotoxicity. Evidence evaluating utilization in the relapsed or refractory setting has been most supported; however, newer trials have also indicated improved survival in the frontline treatment of B-cell ALL. As this therapy is relatively new, the treatment team may include members who are less experienced with the typical treatment course and drug mechanics. This review synthesized available data investigating the effectiveness of blinatumomab effectiveness and its adverse events in addition to providing guidance on safe administration methods utilizing a multidisciplinary healthcare team. When care is coordinated in these settings, serious side effects can be recognized early, allowing for necessary intervention leading to improved quality of life and overall survival. Future research will continue to evaluate blinatumomab in different lines of therapy and expand its way into community settings.
Collapse
Affiliation(s)
- Jeffrey Lantz
- Division of Hematology and Oncology, Department of Medicine, University of Virginia, Charlottesville, VA, USA
| | - Natalie Pham
- Division of Internal Medicine, Department of Medicine, University of Virginia, Charlottesville, VA, USA
| | - Caroline Jones
- Department of Pharmacy, University of Virginia Health System, Charlottesville, VA, USA
| | - Daniel Reed
- Division of Hematology and Oncology, Department of Medicine, University of Virginia, Charlottesville, VA, USA
| | - Firas El Chaer
- Division of Hematology and Oncology, Department of Medicine, University of Virginia, Charlottesville, VA, USA
| | - Michael Keng
- Division of Hematology and Oncology, Department of Medicine, University of Virginia, Charlottesville, VA, USA.
| |
Collapse
|
10
|
Gupta S, Casey J, Lasky J. Case Report: Blinatumomab as upfront consolidation and maintenance therapy in a pediatric patient with high-risk B-cell acute lymphoblastic leukemia. Front Oncol 2023; 13:1246924. [PMID: 38023197 PMCID: PMC10646316 DOI: 10.3389/fonc.2023.1246924] [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: 06/25/2023] [Accepted: 10/16/2023] [Indexed: 12/01/2023] Open
Abstract
Introduction B-cell acute lymphoblastic leukemia (B-ALL) is the most common malignancy in children. The current conventional chemotherapy regimens have high overall survival but with significant short- and long-term toxicities, sometimes requiring delay and termination of chemotherapy. Bispecific T-cell engager antibody blinatumomab has been successful in achieving bone marrow remission and acting as bridging therapy in minimal residual disease (MRD)-positive relapsed adult and pediatric B-ALL patients. Its role as upfront therapy is being explored. Here, we report the first case to our knowledge showing the feasibility, tolerability, and sustained remission using blinatumomab upfront as consolidation and maintenance therapy for 2 years in a pediatric patient with high-risk B-ALL who had significant toxicities with conventional chemotherapy. 'Case presentation An 11-year-old Hispanic girl presented with complaints of fever, abdominal pain, and fatigue. On further evaluation, she had tachycardia, pallor, cervical lymphadenopathy, and pancytopenia. Bone marrow studies confirmed high-risk B-ALL. The patient was started on induction chemotherapy per AALL1131. Her induction course was complicated by syncope, febrile neutropenia, and invasive cryptococcal fungal infection. End-of-induction bone marrow results were MRD negative. Further chemotherapy was withheld due to cardiopulmonary and renal failure, along with ventricular arrhythmias requiring intensive care. The patient received two cycles of blinatumomab as consolidation therapy and then transitioned back to conventional consolidation therapy; however, it was terminated mid-consolidation due to Pseudomonas and Aspergillus sepsis. She was then given blinatumomab maintenance therapy for 2 years and tolerated it well without any irreversible toxicity. She had an episode of Staphylococcus epidermidis sepsis and pneumonia treated by antibiotics and a single episode of a seizure while on blinatumomab therapy. At the time of publication, she is 25 months off treatment and in sustained remission without any further transplant or chemotherapy. She received monthly intravenous immunoglobulin G during the blinatumomab maintenance. Conclusion Blinatumomab given upfront as consolidation and maintenance therapy for 2 years in a pediatric high-risk B-ALL patient with significant toxicities to conventional chemotherapy was feasible and very well tolerated without any irreversible toxicity and led to sustained remission without any bridging transplant or further chemotherapy.
Collapse
Affiliation(s)
- Sumit Gupta
- Department of Pediatric Hematology/Oncology, Cure 4 The Kids, Roseman University of Health Sciences, Las Vegas, NV, United States
- Department of Pediatrics, University of Nevada, Las Vegas, NV, United States
| | - Jessica Casey
- Department of Pediatric Hematology/Oncology, Cure 4 The Kids, Roseman University of Health Sciences, Las Vegas, NV, United States
| | - Joseph Lasky
- Department of Pediatric Hematology/Oncology, Cure 4 The Kids, Roseman University of Health Sciences, Las Vegas, NV, United States
- Department of Pediatrics, University of Nevada, Las Vegas, NV, United States
| |
Collapse
|
11
|
Xie J, Liu S, Zhou M, Wang Y, He H, Xiao P, Hu S, Lu J. Short-course blinatumomab for refractory/relapse precursor B acute lymphoblastic leukemia in children. Front Pediatr 2023; 11:1187607. [PMID: 37601130 PMCID: PMC10437063 DOI: 10.3389/fped.2023.1187607] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/16/2023] [Accepted: 07/20/2023] [Indexed: 08/22/2023] Open
Abstract
Objective To evaluate the clinical efficacy and safety of a short course of blinatumomab in children with refractory or relapsed precursor B-cell acute lymphoblastic leukemia (R/R-BCP-ALL). Methods The clinical data of 33 R/R BCP-ALL children aged 0-18 years who underwent a short course of blinatumomab (14 days) between August 2021 and November 2022 were retrospectively collected and analyzed. Results Among 33 patients with BCP-ALL, 26 achieved complete remission (CR), with a total remission rate of 78.8% (26/33). The duration of remission was approximately 14 days. Of the 7 children without CR, 5 were still in remission at 28 days. In 11 patients with refractory disease and 22 with recurrence, the remission rates were 90.9% (10/11) and 72.7% (16/22), respectively. The overall survival (OS) rates of the 26 patients with CR and seven patients without CR were 96.1% and 57.1% (p = 0.002), respectively, and the disease-free survival (DFS) rates were 96.1% and 42.9% (p < 0.001), respectively. Among the 26 patients with CR, 15 underwent bridging hematopoietic stem cell transplantation (HSCT) and 11 did not receive HSCT; with OS rates of 93.3% and 100% (p = 0.40) and DFS rates of 93.3% and 100% (p = 0.400), respectively. The OS for all patients was 87.9% (29/33) and the DFS was 84.8% (28/33). There were 18 cases (54.5%) of cytokine release syndrome (CRS), 2 cases (6.1%) of severe CRS (all grade 3), 1 case (3.0%) of immune effector cell-associated neurotoxicity syndrome (ICANS), 0 cases (0%) of ICANS ≥ grade 3, and no deaths caused by treatment. Conclusions Short-term follow-up revealed a high R/R BCP-ALL remission rate in children treated with a short course of blinatumomab. The toxicity was low and controllable. No significant short-term survival benefits were observed after bridging HSCT with blinatumomab. In developing countries, a short course of blinatumomab can achieve satisfactory outcomes, while reducing household costs and saving medical resources.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Shaoyan Hu
- Department of Hematology and Oncology, Children’s Hospital of Soochow University, Suzhou, China
| | - Jun Lu
- Department of Hematology and Oncology, Children’s Hospital of Soochow University, Suzhou, China
| |
Collapse
|
12
|
Andreescu M. Risk of Infections Secondary to the Use of Targeted Therapies in Hematological Malignancies. Life (Basel) 2023; 13:1272. [PMID: 37374055 DOI: 10.3390/life13061272] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2023] [Revised: 05/24/2023] [Accepted: 05/27/2023] [Indexed: 06/29/2023] Open
Abstract
Concurrent infections in hematological malignancies (HM) are major contributors to adverse clinical outcomes, including prolonged hospitalization and reduced life expectancy. Individuals diagnosed with HM are particularly susceptible to infectious pathogens due to immunosuppression, which can either be inherent to the hematological disorder or induced by specific therapeutic strategies. Over the years, the treatment paradigm for HM has witnessed a tremendous shift, from broad-spectrum treatment approaches to more specific targeted therapies. At present, the therapeutic landscape of HM is constantly evolving due to the advent of novel targeted therapies and the enhanced utilization of these agents for treatment purposes. By initiating unique molecular pathways, these agents hinder the proliferation of malignant cells, consequently affecting innate and adaptive immunity, which increases the risk of infectious complications. Due to the complexity of novel targeted therapies and their associated risks of infection, it often becomes a daunting task for physicians to maintain updated knowledge in their clinical practice. The situation is further aggravated by the fact that most of the initial clinical trials on targeted therapies provide inadequate information to determine the associated risk of infection. In such a scenario, a cumulative body of evidence is paramount in guiding clinicians regarding the infectious complications that can arise following targeted therapies. In this review, I summarize the recent knowledge on infectious complications arising in the context of targeted therapies for HM.
Collapse
Affiliation(s)
- Mihaela Andreescu
- Department of Clinical Sciences, Hematology, Faculty of Medicine, Titu Maiorescu University of Bucharest, 040051 Bucharest, Romania
- Department of Hematology, Colentina Clinical Hospital, 020125 Bucharest, Romania
| |
Collapse
|
13
|
Pinchinat A, Gupta S, Cooper SL, Rau RE. SOHO State of the Art Updates and Next Questions | Optimal Timing of Blinatumomab for the Treatment of B-Acute Lymphoblastic Leukemia. CLINICAL LYMPHOMA, MYELOMA & LEUKEMIA 2023; 23:159-167. [PMID: 36642665 DOI: 10.1016/j.clml.2022.12.011] [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: 10/20/2022] [Revised: 12/17/2022] [Accepted: 12/19/2022] [Indexed: 12/26/2022]
Abstract
Blinatumomab is a CD19 targeting bi-specific T-cell engager antibody construct developed for the treatment of CD19 expressing B-cell malignancies. Numerous adult and pediatric B-ALL clinical trials have demonstrated blinatumomab's efficacy in the relapse setting as well as in patients with residual disease after upfront chemotherapy. The safety profile of blinatumomab is also favorable, making it a feasible option for most patients. Several key questions remain, including the role of blinatumomab as a replacement for toxic elements of standard chemotherapy regimens in the upfront setting, its role as a bridge to hematopoietic stem cell transplantation, or whether previous blinatumomab impacts the efficacy of subsequent CAR-T cell therapy.
Collapse
Affiliation(s)
- Ashley Pinchinat
- Department of Pediatrics, Division of Pediatric Hematology-Oncology, Hassenfeld Children's Hospital at NYU Langone Health, NYU Grossman School of Medicine, New York, NY
| | - Sumit Gupta
- Division of Haematology/Oncology, Hospital for Sick Children, Toronto, Canada and Faculty of Medicine, University of Toronto, Toronto, Ontario, Canada
| | - Stacy L Cooper
- Department of Oncology, Johns Hopkins University School of Medicine, Baltimore, MD
| | - Rachel E Rau
- Department of Pediatrics, Baylor College of Medicine Texas Children's Hospital, Houston, TX.
| |
Collapse
|
14
|
Kegyes D, Jitaru C, Ghiaur G, Ciurea S, Hoelzer D, Tomuleasa C, Gale RP. Switching from salvage chemotherapy to immunotherapy in adult B-cell acute lymphoblastic leukemia. Blood Rev 2023; 59:101042. [PMID: 36732205 DOI: 10.1016/j.blre.2023.101042] [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: 11/17/2022] [Revised: 12/27/2022] [Accepted: 01/18/2023] [Indexed: 01/21/2023]
Abstract
About one-half of adults with acute B-cell lymphoblastic leukemia (B-ALL) who do not achieve molecular complete remission or who subsequently relapse are not cured by current chemo- or targeted therapies. Previously, the sole therapeutic option for such persons was a hematopoietic stem cell transplant. Recently, several immune therapies including monoclonal antibodies, bispecific T-cell engagers (BiTEs), antibody-drug conjugates (ADCs), and chimeric antigen receptor T-cells (CARs) have been shown safe and effective in this setting. In this manuscript, we summarize data on US FDA-approved immune therapies of advanced adult B-ALL including rituximab, blinatumomab, inotuzumab ozogamicin, tisagenlecleucel and brexucabtagene autoleucel. We consider the results of clinical trials focusing on efficacy, safety, and quality of life (QoL). Real-world evidence is presented as well. We also briefly discuss pharmacodynamics, pharmacokinetics, and pharmacoeconomics followed by risk-benefit analyses. Lastly, we present future directions of immune therapies for advanced B-ALL in adults.
Collapse
Affiliation(s)
- David Kegyes
- Department of Hematology-Medfuture Research Center for Advanced Medicine, Iuliu Hațieganu University of Medicine and Pharmacy Cluj Napoca, Romania; Department of Hematology, Ion Chiricuta Oncology Institute, Cluj Napoca, Romania
| | - Ciprian Jitaru
- Department of Hematology-Medfuture Research Center for Advanced Medicine, Iuliu Hațieganu University of Medicine and Pharmacy Cluj Napoca, Romania; Department of Hematology, Ion Chiricuta Oncology Institute, Cluj Napoca, Romania
| | - Gabriel Ghiaur
- Department of Hematology-Medfuture Research Center for Advanced Medicine, Iuliu Hațieganu University of Medicine and Pharmacy Cluj Napoca, Romania; Department of Leukemia, Johns Hopkins Hospital, Baltimore, MD, USA
| | - Stefan Ciurea
- Department of Stem Cell Transplant and Cellular Therapies, University of California, Irvine, CA, USA
| | - Dieter Hoelzer
- Department of Medicine, Goethe University, Frankfurt, Germany
| | - Ciprian Tomuleasa
- Department of Hematology-Medfuture Research Center for Advanced Medicine, Iuliu Hațieganu University of Medicine and Pharmacy Cluj Napoca, Romania; Department of Hematology, Ion Chiricuta Oncology Institute, Cluj Napoca, Romania.
| | - Robert Peter Gale
- Centre for Haematology, Imperial College of Science, Technology and Medicine, London, UK; Department of Hematologic Oncology, Sun Yat-sen University Cancer Center, Guangzhou, China
| |
Collapse
|
15
|
Barahona-Correa JE, Rueda-Ortiz C, López MJ, Gualtero S, Arevalo-Zambrano M. COVID-19 infection during blinatumomab therapy: Is safety a dilemma? SAGE Open Med Case Rep 2023; 11:2050313X221148548. [PMID: 36643709 PMCID: PMC9834623 DOI: 10.1177/2050313x221148548] [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: 09/29/2022] [Accepted: 12/13/2022] [Indexed: 01/12/2023] Open
Abstract
Patients with acute lymphoblastic leukemia may be particularly vulnerable to SARS-CoV-2 infection and severe illness. The mainstay of current treatment is the use of blinatumomab in patients with refractory or relapsed B-cell precursor acute lymphoblastic leukemia. We discuss the case of a patient with relapsed acute lymphoblastic leukemia who became positive for SARS-CoV-2 during blinatumomab therapy. There are no formal recommendations on the decision to continue, withhold, or delay blinatumomab treatment in these patients. More studies exploring this issue are warranted, as SARS-CoV-2 is expected to be here to stay.
Collapse
Affiliation(s)
- Julián E Barahona-Correa
- Department of Internal Medicine,
Hospital Universitario San Ignacio, Pontificia Universidad Javeriana, Bogota,
Colombia
| | - Camilo Rueda-Ortiz
- Department of Internal Medicine,
Hospital Universitario San Ignacio, Pontificia Universidad Javeriana, Bogota,
Colombia
| | - María-José López
- Department of Internal Medicine,
Hospital Universitario San Ignacio, Pontificia Universidad Javeriana, Bogota,
Colombia,Division of Infectious Diseases,
Hospital Universitario San Ignacio, Bogota, Colombia
| | - Sandra Gualtero
- Department of Internal Medicine,
Hospital Universitario San Ignacio, Pontificia Universidad Javeriana, Bogota,
Colombia,Division of Infectious Diseases,
Hospital Universitario San Ignacio, Bogota, Colombia
| | - Mónica Arevalo-Zambrano
- Department of Internal Medicine,
Hospital Universitario San Ignacio, Pontificia Universidad Javeriana, Bogota,
Colombia,Department of Hematology, Hospital
Universitario San Ignacio, Bogota, Colombia,Mónica Arevalo-Zambrano, Carrera 7 #,
42-65, Bogota, Colombia.
| |
Collapse
|
16
|
Sim B, Ng JY, Teh BW, Talaulikar D. Immunoglobulin replacement in hematological malignancies: a focus on evidence, alternatives, dosing strategy, and cessation rule. Leuk Lymphoma 2023; 64:18-29. [PMID: 36218218 DOI: 10.1080/10428194.2022.2131424] [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: 11/06/2022]
Abstract
Acquired hypogammaglobulinemia or secondary immunodeficiency (SID) occurs commonly in hematological malignancies with increasing incidence and complexity in the era of modern therapies. Despite current practice of immunoglobulin replacement (IgRT) in SID, the evidence is lacking, especially for newer treatments. We discuss the current evidence for IgRT in various disease groups including issues, such as actual or ideal body weight (IBW)-based dosing, length of treatment, antibiotic prophylaxis, and vaccination. Incidence of SID with newer treatment is lacking. While there is a trend toward decreased respiratory infections and hospitalizations with IgRT, this is not consistent across all disease course or treatment groups. Dosing and indications for cessation of IgRT are also inadequately characterized. Further randomized controlled trials (RCTs) and observational studies are required to assess the optimal indications, timing, and duration of IgRT to improve the efficacy, safety, and cost-effectiveness. Assessment of alternative and adjunctive therapies, such as vaccination and antibiotic prophylaxis could also improve the outcomes and costs.
Collapse
Affiliation(s)
- Beatrice Sim
- Department of Infectious Diseases, Peter MacCallum Cancer Centre, Melbourne, Australia
| | - Jun Yen Ng
- Department of Hematology, ACT Pathology, Canberra Hospital, Canberra, Australia
| | - Benjamin W Teh
- Department of Infectious Diseases, Peter MacCallum Cancer Centre, Melbourne, Australia.,Sir Peter MacCallum Department of Oncology, University of Melbourne, Melbourne, Australia
| | - Dipti Talaulikar
- Department of Hematology, ACT Pathology, Canberra Hospital, Canberra, Australia.,College of Health and Medicine, Australian National University, Canberra, Australia
| |
Collapse
|
17
|
Joseph A, Lafarge A, Azoulay E, Zafrani L. Acute Kidney Injury in Cancer Immunotherapy Recipients. Cells 2022; 11:cells11243991. [PMID: 36552755 PMCID: PMC9776910 DOI: 10.3390/cells11243991] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2022] [Revised: 12/02/2022] [Accepted: 12/08/2022] [Indexed: 12/14/2022] Open
Abstract
Cancer immunotherapy has now entered clinical practice and has reshaped the standard of care for many cancer patients. With these new strategies, specific toxicities have emerged, and renal side effects have been described. In this review, we will describe the causes of acute kidney injury in CAR T cell, immune checkpoint inhibitors and other cancer immuno-therapy recipients. CAR T cell therapy and bispecific T cell engaging antibodies can lead to acute kidney injury as a consequence of cytokine release syndrome, tumor lysis syndrome, sepsis or specific CAR T cell infiltration. Immune checkpoint blockade most often results in acute tubular interstitial nephritis, but glomerular diseases have also been described. Although the pathophysiology remains mostly elusive, we will describe the mechanisms of renal damage in these contexts, its prognosis and treatment. As the place of immunotherapy in the anti-cancer armamentarium is exponentially increasing, close collaboration between nephrologists and oncologists is of utmost importance to provide the best standard of care for these patients.
Collapse
|
18
|
Joseph A, Lafarge A, Mabrouki A, Abdel-Nabey M, Binois Y, Younan R, Azoulay E. Severe infections in recipients of cancer immunotherapy: what intensivists need to know. Curr Opin Crit Care 2022; 28:540-550. [PMID: 35950720 DOI: 10.1097/mcc.0000000000000978] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
PURPOSE OF REVIEW Given the increased number of cancer patients admitted in the ICU and the growing importance of immunotherapy in their therapeutic arsenal, intensivists will be increasingly confronted to patients treated with immunotherapies who will present with complications, infectious and immunologic. RECENT FINDINGS Apart from their specific immunologic toxicities, cancer immunotherapy recipients also have specific immune dysfunction and face increased infectious risks that may lead to intensive care unit admission. SUMMARY Chimeric antigen receptor T-cell therapy is associated with profound immunosuppression and the risks of bacterial, fungal and viral infections vary according to the time since infusion.Immune checkpoint blockers are associated with an overall favorable safety profile but associations of checkpoint blockers and corticosteroids and immunosuppressive drugs prescribed to treat immune-related adverse events are associated with increased risks of bacterial and fungal infections.The T-cell engaging bispecific therapy blinatumomab causes profound B-cell aplasia, hypogammaglobulinemia and neutropenia, but seems to be associated with fewer infectious adverse events compared with standard intensive chemotherapy.Lastly, intravesical administration of Bacillus Calmette-Guérin (BCG) can lead to disseminated BCGitis and severe sepsis requiring a specific antibiotherapy, often associated with corticosteroid treatment.
Collapse
Affiliation(s)
- Adrien Joseph
- Medical Intensive Care Unit, Saint-Louis Teaching Hospital, Public Assistance Hospitals of Paris, Paris, France
| | | | | | | | | | | | | |
Collapse
|
19
|
Essa MF, Abdellatif R, Elimam N, Ballourah W, Alsudairy R, Alkaiyat M, Alsultan A, Jastaniah W. Outcomes of blinatumomab based therapy in children with relapsed, persistent, or refractory acute lymphoblastic leukemia: a multicenter study focusing on predictors of response and post-treatment immunoglobulin production. Pediatr Hematol Oncol 2022; 39:613-628. [PMID: 36200934 DOI: 10.1080/08880018.2022.2049936] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 10/18/2022]
Abstract
The management of Refractory/Relapsed B-cell Acute Lymphoblastic Leukemia (R/R ALL) remains challenging. Incorporating blinatumomab in R/R ALL treatment has shown encouraging results. We describe the outcome and predictors of response in children receiving blinatumomab as a bridge to definitive therapy. Immunoglobulin (Ig) G and viral serology before and after therapy were evaluated. Thirty-three patients that failed standard first-line treatments due to relapsed ALL (n = 22), persistent minimal residual disease (MRD) (n = 8), or refractory disease (n = 3) received blinatumomab. Grade 2 toxicity occurred in 27.2% of patients. MRD remission (<0.01%) was achieved in 72.7% of patients. Pre-blinatumomab absolute lymphocyte count (ALC) and MRD/ALC ratio significantly associated with MRD-response. Patients with t(1;19) translocation had lower response rate, compared to all other cytogenetic categories (p = 0.013). One-year event-free survival (EFS) and overall survival (OS) were 69.2% and 79.7%, respectively. Analysis of OS and EFS showed pre-blinatumomab MRD level, ALC, MRD/ALC ratio, t(1;19), and post-blinatumomab MRD remission associated with survival. Following blinatumomab, 83% (15/18) of tested patients had low IgG levels. IgG seronegative status was observed in 83% (12/15) for varicella zoster, 35% (6/17) for herpes zoster, 18% (3/17) for cytomegalovirus, and 26% (5/17) for Epstein Barr virus. Blinatumomab produced encouraging results in children with R/R ALL and low disease burden bridging to definitive therapy. Incorporating baseline genetics and biomarkers may help identify subgroups likely to be responsive/resistant to therapy. Viral serological testing pre- and post-blinatumomab is recommended to optimize supportive and preemptive therapy.Supplemental data for this article is available online at https://doi.org/10.1080/08880018.2022.2049936 .
Collapse
Affiliation(s)
- Mohammed F Essa
- Department of Pediatric Hematology/Oncology, King Abdullah Specialist Children's Hospital, Ministry of National Guard Health Affairs, Riyadh, Saudi Arabia.,College of Medicine, King Saud bin Abdulaziz University for Health Sciences, Riyadh, Saudi Arabia.,King Abdullah International Medical Research Center, National Guard Health Affairs, Riyadh, Saudi Arabia
| | - Reem Abdellatif
- Department of Pediatric Hematology/Oncology, King Abdullah Specialist Children's Hospital, Ministry of National Guard Health Affairs, Riyadh, Saudi Arabia
| | - Naglla Elimam
- King Abdullah International Medical Research Center, National Guard Health Affairs, Riyadh, Saudi Arabia.,Department of Oncology, Princess Noorah Oncology Center, Ministry of National Guard Health Affairs, Jeddah, Saudi Arabia
| | - Waleed Ballourah
- Department of Pediatric Hematology/Oncology, Comprehensive Cancer Center, King Fahd Medical City, Riyadh, Saudi Arabia
| | - Reem Alsudairy
- Department of Pediatric Hematology/Oncology, King Abdullah Specialist Children's Hospital, Ministry of National Guard Health Affairs, Riyadh, Saudi Arabia
| | - Mohammad Alkaiyat
- Department of Oncology, King Abdulaziz Medical City, Ministry of National Guard Health Affairs, Riyadh, Saudi Arabia
| | - Abdulrahman Alsultan
- Department of Oncology, King Abdulaziz Medical City, Ministry of National Guard Health Affairs, Riyadh, Saudi Arabia.,Department of Pediatrics, College of Medicine, King Saud University, Riyadh, Saudi Arabia.,Oncology Center, King Saud University Medical City, Riyadh, Saudi Arabia
| | - Wasil Jastaniah
- Department of Oncology, King Faisal Specialist Hospital & Research Center (Gen.Org.), Jeddah, Saudi Arabia
| |
Collapse
|
20
|
Podpeskar A, Crazzolara R, Kropshofer G, Obexer P, Rabensteiner E, Michel M, Salvador C. Supportive methods for childhood acute lymphoblastic leukemia then and now: A compilation for clinical practice. Front Pediatr 2022; 10:980234. [PMID: 36172391 PMCID: PMC9510731 DOI: 10.3389/fped.2022.980234] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/28/2022] [Accepted: 08/15/2022] [Indexed: 12/02/2022] Open
Abstract
Survival of childhood acute lymphoblastic leukemia has significantly improved over the past decades. In the early years of chemotherapeutic development, improvement in survival rates could be attained only by increasing the cytostatic dose, also by modulation of the frequency and combination of chemotherapeutic agents associated with severe short- and long-time side-effects and toxicity in a developing child's organism. Years later, new treatment options have yielded promising results through targeted immune and molecular drugs, especially in relapsed and refractory leukemia, and are continuously added to conventional therapy or even replace first-line treatment. Compared to conventional strategies, these new therapies have different side-effects, requiring special supportive measures. Supportive treatment includes the prevention of serious acute and sometimes life-threatening events as well as managing therapy-related long-term side-effects and preemptive treatment of complications and is thus mandatory for successful oncological therapy. Inadequate supportive therapy is still one of the main causes of treatment failure, mortality, poor quality of life, and unsatisfactory long-term outcome in children with acute lymphoblastic leukemia. But nowadays it is a challenge to find a way through the flood of supportive recommendations and guidelines that are available in the literature. Furthermore, the development of new therapies for childhood leukemia has changed the range of supportive methods and must be observed in addition to conventional recommendations. This review aims to provide a clear and recent compilation of the most important supportive methods in the field of childhood leukemia, based on conventional regimes as well as the most promising new therapeutic approaches to date.
Collapse
Affiliation(s)
- Alexandra Podpeskar
- Division of Hematology and Oncology, Department of Pediatrics I, Medical University of Innsbruck, Innsbruck, Austria
| | - Roman Crazzolara
- Division of Hematology and Oncology, Department of Pediatrics I, Medical University of Innsbruck, Innsbruck, Austria
| | - Gabriele Kropshofer
- Division of Hematology and Oncology, Department of Pediatrics I, Medical University of Innsbruck, Innsbruck, Austria
| | - Petra Obexer
- Department of Pediatrics II, Medical University of Innsbruck, Innsbruck, Austria
| | - Evelyn Rabensteiner
- Division of Hematology and Oncology, Department of Pediatrics I, Medical University of Innsbruck, Innsbruck, Austria
| | - Miriam Michel
- Division of Cardiology, Department of Pediatrics III, Medical University of Innsbruck, Innsbruck, Austria
| | - Christina Salvador
- Division of Hematology and Oncology, Department of Pediatrics I, Medical University of Innsbruck, Innsbruck, Austria
| |
Collapse
|
21
|
Mocquot P, Mossazadeh Y, Lapierre L, Pineau F, Despas F. The pharmacology of blinatumomab: state of the art on pharmacodynamics, pharmacokinetics, adverse drug reactions and evaluation in clinical trials. J Clin Pharm Ther 2022; 47:1337-1351. [PMID: 35906791 PMCID: PMC9796714 DOI: 10.1111/jcpt.13741] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2022] [Revised: 06/27/2022] [Accepted: 06/29/2022] [Indexed: 01/07/2023]
Abstract
WHAT IS KNOWN AND OBJECTIVE Bispecific drugs (BDs) belong to the family of immunotherapies along with checkpoint inhibitors and CAR-T cells. In the field of oncology, BDs are designed to simultaneously bind a tumour antigen on the one side and an antigen present on the surface of effector cells on the other. This review summarizes the information available to date on the first marketed BiTE-format bispecific antibody, blinatumomab BLINCYTO® in acute lymphoblastic leukaemia. METHODS A literature search was conducted in the PubMed database by including studies published in English using the term blinatumomab. Furthermore, bibliographies of selected references were also evaluated for relevant articles. Clinical trial (CT) data were retrieved from clinicaltrials.gov (ongoing trials, adverse events [AEs]) and global pharmacovigilance data were retrieved from VigiBase®. RESULTS AND DISCUSSION Blinatumomab is a fusion protein which consists of two single-chain variable fragments arranged in tandem: the first binds the CD19 surface antigen of all B cells and the second targets the CD3 antigen of T cells. Binding of blinatumomab to B and T cells induces apoptosis of B cells after secretion of granzymes and perforins by T cells. T-cell activation results in secretion of pro-inflammatory cytokines and upregulation of activation markers and adhesion molecules on the surface of T cells. The major CTs that led to an indication show increased overall survival with blinatumomab with better efficacy in patients in haematological remission with minimal residual disease ≥10-3 . The major AEs are cytokine release syndrome, neurotoxicity and hypogammaglobulinemia. The three most frequent system organ classes in CTs are haematological, gastrointestinal and general disorders. These results are also found in VigiBase® but neurological disorders and infections appear more frequently in real life. WHAT IS NEW AND CONCLUSION This review summarizes the current knowledge of blinatumomab in the literature. The subject of many CTs is to improve the route of administration and expand the indications for treatment.
Collapse
Affiliation(s)
- Pauline Mocquot
- Département de Pharmacologie Médicale, CHU de ToulouseUniversité Toulouse III ‐ Paul SabatierToulouseFrance
| | - Yasmine Mossazadeh
- Département de Pharmacologie Médicale, CHU de ToulouseUniversité Toulouse III ‐ Paul SabatierToulouseFrance
| | - Léopoldine Lapierre
- Département d'Hématologie et de Médecine InterneInstitut Universitaire du Cancer‐Oncopole, CHU de ToulouseToulouseFrance
| | - Fanny Pineau
- Département d'Hématologie et de Médecine InterneInstitut Universitaire du Cancer‐Oncopole, CHU de ToulouseToulouseFrance
| | - Fabien Despas
- Département de Pharmacologie Médicale, CHU de ToulouseUniversité Toulouse III ‐ Paul SabatierToulouseFrance,Université Toulouse III ‐ Paul SabatierToulouseFrance,INSERM CIC1436 CIC ToulouseFrance
| |
Collapse
|
22
|
Paredes-Amaya CC, Matta-Cortes L, Zea-Vera AF. Severe coronavirus HCoV-NL63 pneumonia in a patient receiving blinatumomab with secondary antibody deficiency in COVID-19 times. Germs 2022; 12:292-297. [PMID: 36504603 PMCID: PMC9719380 DOI: 10.18683/germs.2022.1331] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2021] [Revised: 03/30/2022] [Accepted: 04/17/2022] [Indexed: 12/15/2022]
Abstract
Introduction Human coronavirus NL63 (HCoV-NL63) is one of four common human respiratory coronaviruses. It causes lower respiratory tract infections in young children, elderly and immunosuppressed people, which could result in fatal outcomes. In this time of pandemic, we want to highlight the importance of other coronaviruses infection besides SARS-CoV-2, especially in a patient with underlying conditions like acute lymphoblastic leukemia, receiving immunosuppressive therapy that could result in humoral secondary immunodeficiencies. Case report We present the case of a 44-year-old Colombian man with acute lymphoblastic leukemia who developed HCoV-NL63 pulmonary infection after the first month of treatment with blinatumomab complicated with severe secondary hypogammaglobulinemia. HCoV-NL63 was detected by multiplex PCR, and HCoV-NL63 viral pneumonia was diagnosed. Hypogammaglobulinemia was studied by determining serum immunoglobulins levels and protein electrophoresis. The treatment consisted of supportive therapy and replacement with intravenous immunoglobulins. After therapy, the patient improved his oxygenation, and the infection was resolved in a few days. Conclusions This case highlights the relevance of other coronaviruses infections besides SARS-CoV-2 in patients receiving immunosuppressive therapy who develop secondary antibody deficiency, and the importance of replacement therapy with intravenous immunoglobulins at early stage of infection with HCoV-NL63.
Collapse
Affiliation(s)
- Claudia C. Paredes-Amaya
- PhD, Departamento de Microbiología, Facultad de Salud, Universidad del Valle, CP 760043, Cali, Colombia
| | - Lorena Matta-Cortes
- MD, MSc, Departamento de Medicina Interna, Facultad de Salud, Universidad del Valle, CP 760043, Cali, Colombia
| | - Andrés Felipe Zea-Vera
- MD, PhD, Departamento de Microbiología, Facultad de Salud, Universidad del Valle, Calle 4b 36-00 Edificio 120, CP 760043, Cali, Colombia,Corresponding author: Andrés Felipe Zea-Vera,
| |
Collapse
|
23
|
Wo S, Levavi H, Mascarenhas J, Kremyanskaya M, Navada S, Bar-Natan M, Kim SS. Immunoglobulin repletion during blinatumomab therapy does not reduce the rate of secondary hypogammaglobulinemia and associated infectious risk. Blood Res 2022; 57:135-143. [PMID: 35551109 PMCID: PMC9242831 DOI: 10.5045/br.2022.2021163] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2021] [Revised: 01/09/2022] [Accepted: 04/18/2022] [Indexed: 11/17/2022] Open
Abstract
Background Blinatumomab has demonstrated efficacy in minimal residual disease (MRD) positive and relapsed/refractory B-cell acute lymphoblastic leukemia (B-ALL) by inciting rapid and sustained B-cell depletion. Methods Owing to its effect on B-cells, blinatumomab is associated with a higher rate of secondary hypogammaglobulinemia compared to chemotherapy. To mitigate blinatumomab-induced hypogammaglobulinemia, patients were pre-emptively repleted with intravenous immune globulin (IVIG) during blinatumomab therapy. In this retrospective study, we compared outcomes of 23 blinatumomab treated adults with ALL. Seventeen patients routinely received IVIG and 6 patients were in the control cohort. Results Our findings demonstrated no difference between the two cohorts in immunoglobulin G (IgG) nadir (338 mg/dL vs. 337 mg/dL, P=0.641), days to IgG nadir (120.5 vs. 85.5 days, P=0.13), infection rate (82.4% vs. 66.7%, P=0.58), infections requiring ICU admission (23.5% vs. 16.7%, P=1), and infection related mortality (17.6% vs. 16.7%, P=1). Conclusion Pre-emptive IVIG repletion during blinatumomab did not prevent hypogammaglobulinemia and associated infection risk.
Collapse
Affiliation(s)
- Stephanie Wo
- Department of Pharmacy, The Mount Sinai Hospital, New York, NY, USA
| | - Hannah Levavi
- Division of Hematology/Oncology, Icahn School of Medicine at The Mount Sinai Hospital, New York, NY, USA
| | - John Mascarenhas
- Division of Hematology/Oncology, Icahn School of Medicine at The Mount Sinai Hospital, New York, NY, USA
| | - Marina Kremyanskaya
- Division of Hematology/Oncology, Icahn School of Medicine at The Mount Sinai Hospital, New York, NY, USA
| | - Shyamala Navada
- Division of Hematology/Oncology, Icahn School of Medicine at The Mount Sinai Hospital, New York, NY, USA
| | - Michal Bar-Natan
- Division of Hematology/Oncology, Icahn School of Medicine at The Mount Sinai Hospital, New York, NY, USA
| | - Sara S Kim
- Department of Pharmacy, The Mount Sinai Hospital, New York, NY, USA
| |
Collapse
|
24
|
Effects of B-Cell Lymphoma on the Immune System and Immune Recovery after Treatment: The Paradigm of Targeted Therapy. Int J Mol Sci 2022; 23:ijms23063368. [PMID: 35328789 PMCID: PMC8952275 DOI: 10.3390/ijms23063368] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2022] [Revised: 03/07/2022] [Accepted: 03/09/2022] [Indexed: 12/10/2022] Open
Abstract
B-cell lymphoma and lymphoproliferative diseases represent a heterogeneous and complex group of neoplasms that are accompanied by a broad range of immune regulatory disorder phenotypes. Clinical features of autoimmunity, hyperinflammation, immunodeficiency and infection can variously dominate, depending on the immune pathway most involved. Immunological imbalance can play a role in lymphomagenesis, also supporting the progression of the disease, while on the other hand, lymphoma acts on the immune system to weaken immunosurveillance and facilitate immunoevasion. Therefore, the modulation of immunity can have a profound effect on disease progression or resolution, which makes the immune system a critical target for new therapies. In the current therapeutic scenario enriched by chemo-free regimens, it is important to establish the effect of various drugs on the disease, as well as on the restoration of immune functions. In fact, treatment of B-cell lymphoma with passive immunotherapy that targets tumor cells or targets the tumor microenvironment, together with adoptive immunotherapy, is becoming more frequent. The aim of this review is to report relevant data on the evolution of the immune system during and after treatment with targeted therapy of B-cell lymphomas.
Collapse
|
25
|
Little JS, Weiss ZF, Hammond SP. Invasive Fungal Infections and Targeted Therapies in Hematological Malignancies. J Fungi (Basel) 2021; 7:1058. [PMID: 34947040 PMCID: PMC8706272 DOI: 10.3390/jof7121058] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2021] [Revised: 12/03/2021] [Accepted: 12/06/2021] [Indexed: 01/02/2023] Open
Abstract
The use of targeted biologic therapies for hematological malignancies has greatly expanded in recent years. These agents act upon specific molecular pathways in order to target malignant cells but frequently have broader effects involving both innate and adaptive immunity. Patients with hematological malignancies have unique risk factors for infection, including immune dysregulation related to their underlying disease and sequelae of prior treatment regimens. Determining the individual risk of infection related to any novel agent is challenging in this setting. Invasive fungal infections (IFIs) represent one of the most morbid infectious complications observed in hematological malignancy. In recent years, growing evidence suggests that certain small molecule inhibitors, such as BTK inhibitors and PI3K inhibitors, may cause an increased risk of IFI in certain patients. It is imperative to better understand the impact that novel targeted therapies might have on the development of IFIs in this high-risk patient population.
Collapse
Affiliation(s)
- Jessica S. Little
- Division of Infectious Diseases, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA 02115, USA;
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA 02215, USA;
| | - Zoe F. Weiss
- Division of Infectious Diseases, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA 02115, USA;
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA 02215, USA;
| | - Sarah P. Hammond
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA 02215, USA;
- Division of Infectious Diseases, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02114, USA
- Division of Hematology and Oncology, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02114, USA
| |
Collapse
|
26
|
Impact of Blinatumomab Treatment on Bone Marrow Function in Patients with Relapsed/Refractory B-Cell Precursor Acute Lymphoblastic Leukemia. Cancers (Basel) 2021; 13:cancers13225607. [PMID: 34830762 PMCID: PMC8616108 DOI: 10.3390/cancers13225607] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2021] [Revised: 11/02/2021] [Accepted: 11/06/2021] [Indexed: 12/12/2022] Open
Abstract
Simple Summary Myelosuppression is a side effect of chemotherapy, in which the production of red and white blood cells and platelets is reduced, increasing risk of infections. Blinatumomab, a bispecific T-cell engager (BiTE®) molecule, is a novel anticancer drug that kills acute lymphoblastic leukemia (ALL) cells while sparing majority of the normal bone marrow cells. Results from our study, which compared the effects of blinatumomab treatment with chemotherapy on bone marrow function in a large number of patients with ALL, indicated that the decrease in blood cell counts was more severe and lasted longer after chemotherapy compared with blinatumomab treatment, in which the effects were transient. Survival in patients treated with blinatumomab achieving complete remission was more similar between those with incomplete recovery of blood cell counts versus those with complete blood cell counts than the corresponding survival outcomes seen with chemotherapy. In conclusion, blinatumomab treatment caused transient myelosuppression when compared with chemotherapy. Abstract Association of blinatumomab treatment with myelosuppression was examined in this study. Peripheral blood counts were assessed prior to, during, and after blinatumomab treatment in patients with relapsed/refractory Philadelphia chromosome-negative (Ph−) B-cell precursor (BCP) acute lymphoblastic leukemia (ALL; n = 267) and Ph+ BCP-ALL (n = 45) from the TOWER and ALCANTARA studies, respectively, or chemotherapy in patients with Ph− BCP-ALL (n = 109) from the TOWER study; all the patients with relapsed/refractory BCP-ALL and responders achieving complete remission (CR) or CR with partial/incomplete hematological recovery (CRh/CRi) were evaluated. Event-free survival (EFS) and overall survival (OS) were assessed in patients achieving CR and CRh/CRi. Median leukocyte, neutrophil, and platelet counts increased during two blinatumomab cycles but remained low longer after chemotherapy. Among the responders, there was a trend that a greater proportion of patients achieved CR with blinatumomab (Ph−, 76.5%; Ph+, 77.8%) versus with chemotherapy (Ph−, 63.6%). In the TOWER study, the survival prognosis for patients achieving CRh/CRi versus CR with blinatumomab was more similar (median OS, 11.9 (95% CI, 3.9–not estimable (NE)) vs. 15.0 (95% CI, 10.4–NE) months, p = 0.062) than with chemotherapy (5.2 (95% CI, 1.6–NE) vs. 18.9 (95% CI, 9.3–NE) months, p = 0.013). Blinatumomab treatment, with only temporary and transient myelosuppression, resulted in a greater survival benefit than chemotherapy.
Collapse
|
27
|
Secondary Dysgammaglobulinemia in Children with Hematological Malignancies Treated with Targeted Therapies. Paediatr Drugs 2021; 23:445-455. [PMID: 34292515 DOI: 10.1007/s40272-021-00461-3] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 07/06/2021] [Indexed: 10/20/2022]
Abstract
Targeted therapies have emerged as innovative treatments for patients whose disease does not respond to conventional chemotherapy, and their use has widely expanded in the field of pediatric hematologic malignancies in the last decade. While they carry the promise of improved disease control and survival and are currently investigated in first-line treatment protocols for patients with poor prognostic markers, they are associated with a considerable incidence of specific toxicities, including cytokine-release syndrome, neurotoxicity, hepatotoxicity, nephrotoxicity, cardiotoxicity, endocrine adverse events, and infectious complications. Iatrogenic or secondary dysgammaglobulinemia is a main consequence of targeted therapies using monoclonal antibodies and other antibody-derived treatments that target specific antigens on lymphoid cells (blinatumomab, inotuzumab ozogamicin, rituximab), chimeric antigen receptor T cells, tyrosine kinase inhibitors (imatinib, dasatinib, nilotinib) and, to a lesser extent, checkpoint inhibitors (pembrolizumab, nivolumab). This review discusses the diagnosis and incidence of secondary or iatrogenic dysgammaglobulinemia in children treated with targeted therapies for leukemias and lymphomas, and options for monitoring and treatment.
Collapse
|
28
|
Abstract
This article has a companion Point by Molina and Shah.
Collapse
|
29
|
Kyriakidis I, Vasileiou E, Rossig C, Roilides E, Groll AH, Tragiannidis A. Invasive Fungal Diseases in Children with Hematological Malignancies Treated with Therapies That Target Cell Surface Antigens: Monoclonal Antibodies, Immune Checkpoint Inhibitors and CAR T-Cell Therapies. J Fungi (Basel) 2021; 7:186. [PMID: 33807678 PMCID: PMC7999508 DOI: 10.3390/jof7030186] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2021] [Revised: 02/27/2021] [Accepted: 03/02/2021] [Indexed: 12/12/2022] Open
Abstract
Since 1985 when the first agent targeting antigens on the surface of lymphocytes was approved (muromonab-CD3), a multitude of such therapies have been used in children with hematologic malignancies. A detailed literature review until January 2021 was conducted regarding pediatric patient populations treated with agents that target CD2 (alefacept), CD3 (bispecific T-cell engager [BiTE] blinatumomab), CD19 (denintuzumab mafodotin, B43, BiTEs blinatumomab and DT2219ARL, the immunotoxin combotox, and chimeric antigen receptor [CAR] T-cell therapies tisagenlecleucel and axicabtagene ciloleucel), CD20 (rituximab and biosimilars, 90Y-ibritumomab tiuxetan, ofatumumab, and obinutuzumab), CD22 (epratuzumab, inotuzumab ozogamicin, moxetumomab pasudotox, BiTE DT2219ARL, and the immunotoxin combotox), CD25 (basiliximab and inolimomab), CD30 (brentuximab vedotin and iratumumab), CD33 (gemtuzumab ozogamicin), CD38 (daratumumab and isatuximab), CD52 (alemtuzumab), CD66b (90Y-labelled BW 250/183), CD248 (ontuxizumab) and immune checkpoint inhibitors against CTLA-4 (CD152; abatacept, ipilimumab and tremelimumab) or with PD-1/PD-L1 blockade (CD279/CD274; atezolizumab, avelumab, camrelizumab, durvalumab, nivolumab and pembrolizumab). The aim of this narrative review is to describe treatment-related invasive fungal diseases (IFDs) of each category of agents. IFDs are very common in patients under blinatumomab, inotuzumab ozogamicin, basiliximab, gemtuzumab ozogamicin, alemtuzumab, and tisagenlecleucel and uncommon in patients treated with moxetumomab pasudotox, brentuximab vedotin, abatacept, ipilimumab, pembrolizumab and avelumab. Although this new era of precision medicine shows promising outcomes of targeted therapies in children with leukemia or lymphoma, the results of this review stress the necessity for ongoing surveillance and suggest the need for antifungal prophylaxis in cases where IFDs are very common complications.
Collapse
Affiliation(s)
- Ioannis Kyriakidis
- Pediatric and Adolescent Hematology-Oncology Unit, 2nd Department of Pediatrics, Faculty of Health Sciences, School of Medicine, Aristotle University of Thessaloniki, AHEPA Hospital, 54636 Thessaloniki, Greece; (I.K.); (E.V.)
| | - Eleni Vasileiou
- Pediatric and Adolescent Hematology-Oncology Unit, 2nd Department of Pediatrics, Faculty of Health Sciences, School of Medicine, Aristotle University of Thessaloniki, AHEPA Hospital, 54636 Thessaloniki, Greece; (I.K.); (E.V.)
| | - Claudia Rossig
- Department of Pediatric Hematology and Oncology, University Children’s Hospital Münster, D-48149 Münster, Germany;
| | - Emmanuel Roilides
- Infectious Diseases Unit, Basic and Translational Research Unit, Special Unit for Biomedical Research and Education, 3rd Department of Pediatrics, Faculty of Health Sciences, School of Medicine, Aristotle University of Thessaloniki, Hippokration General Hospital, 54642 Thessaloniki, Greece;
| | - Andreas H. Groll
- Center for Bone Marrow Transplantation and Department of Pediatric Hematology and Oncology, Infectious Disease Research Program, University Children’s Hospital Münster, D-48149 Münster, Germany;
| | - Athanasios Tragiannidis
- Pediatric and Adolescent Hematology-Oncology Unit, 2nd Department of Pediatrics, Faculty of Health Sciences, School of Medicine, Aristotle University of Thessaloniki, AHEPA Hospital, 54636 Thessaloniki, Greece; (I.K.); (E.V.)
- Center for Bone Marrow Transplantation and Department of Pediatric Hematology and Oncology, Infectious Disease Research Program, University Children’s Hospital Münster, D-48149 Münster, Germany;
| |
Collapse
|
30
|
Abstract
PURPOSE OF REVIEW Tremendous advances have been made in the treatment armamentarium for acute lymphoblastic leukemia in recent years, which have substantially improved outcomes for these patients. At the same time, unique toxicities have emerged, and without early intervention, are life-threatening. This article will review the novel therapies in acute leukemias and highlight the clinically relevant supportive care advances. RECENT FINDINGS The American Society for Transplantation and Cellular Therapy (ASTCT) has put forth the most recent recommendations in managing the cytokine release syndrome and neurotoxicity after chimeric antigen receptor T cells (CAR-T) and blinatumomab. The hepatic injury incurred by inotuzumab, and the vascular toxicity of tyrosine kinase inhibitors, other relatively novel agents, require subspecialist intervention and multidisciplinary care. Asparaginase, a long-established and key element of pediatric regimens, has made a comeback in the young adult leukemia population. Updated guidelines have been outlined for management of asparaginase thrombotic complications. Lastly, although there have been few changes in the applications of growth factor, antimicrobial prophylaxis, and management of neuropathy, these encompass exceedingly important aspects of care. While the rapidly changing treatment paradigms for acute lymphoblastic leukemia have transformed leukemia-specific outcomes, treatment emergent toxicities have forced much necessary attention to better definitions of these toxicities and on improving supportive care guidelines in acute lymphoblastic leukemia.
Collapse
|
31
|
McMasters M, Blair BM, Lazarus HM, Alonso CD. Casting a wider protective net: Anti-infective vaccine strategies for patients with hematologic malignancy and blood and marrow transplantation. Blood Rev 2020; 47:100779. [PMID: 33223246 DOI: 10.1016/j.blre.2020.100779] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2020] [Revised: 10/29/2020] [Accepted: 11/04/2020] [Indexed: 02/07/2023]
Abstract
Patients who have hematologic malignancies are at high risk for infections but vaccinations may be effective prophylaxis. The increased infection risk derives from immune defects secondary to malignancy, the classic example being CLL, and chemotherapies and immunotherapy used to treat the malignancies. Therapy of hematologic malignancies is being revolutionized by introduction of novel targeted agents and immunomodulatory medications, improving the survival of patients. At the same time those agents uniquely change the infection risk and response to immunizations. This review will summarize current vaccine recommendations for patients with hematologic malignancies including patients who undergo hematopoietic cell transplant.
Collapse
Affiliation(s)
- Malgorzata McMasters
- Division of Hematologic Malignancy and Bone Marrow Transplant, Beth Israel Deaconess Medical Center, 330 Brookline Ave, Boston, MA 02215, USA; Harvard Medical School, Boston, MA, USA
| | - Barbra M Blair
- Harvard Medical School, Boston, MA, USA; Division of Infectious Diseases, Beth Israel Deaconess Medical Center, 110 Francis Street, Suite GB, Boston, MA 02215, USA
| | - Hillard M Lazarus
- Department of Medicine, Case Western Reserve University, Cleveland, OH 44106, USA
| | - Carolyn D Alonso
- Harvard Medical School, Boston, MA, USA; Division of Infectious Diseases, Beth Israel Deaconess Medical Center, 110 Francis Street, Suite GB, Boston, MA 02215, USA.
| |
Collapse
|
32
|
Conde-Royo D, Juárez-Salcedo LM, Dalia S. Management of adverse effects of new monoclonal antibody treatments in acute lymphoblastic leukemia. Drugs Context 2020; 9:dic-2020-7-2. [PMID: 33110433 PMCID: PMC7560100 DOI: 10.7573/dic.2020-7-2] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2020] [Revised: 09/15/2020] [Accepted: 09/16/2020] [Indexed: 02/07/2023] Open
Abstract
Therapeutic options for relapsed/refractory B-cell acute lymphoblastic leukemia have evolved in the past few years. The FDA has approved three novel therapies for this disease: inotuzumab ozogamicin (an anti-CD22 antibody–drug conjugate), blinatumomab (a bispecific T-cell engager), and chimeric antigen receptor T-cell therapy. Although these novel immunotherapies have revolutionized the therapeutic landscape, it is important to understand the crucial aspects of administration, especially toxicity. In this article, we review the unique toxicities and adverse effects of blinatumomab and inotuzumab ozogamicin and provide recommendations for prevention of adverse effects as well as the management options for each medication.
Collapse
Affiliation(s)
- Diego Conde-Royo
- Hematology Department, Principe de Asturias General Hospital, Madrid, Spain
| | | | - Samir Dalia
- Hematology/Oncology Department, Mercy Clinic Oncology and Hematology - Joplin, MO, USA
| |
Collapse
|
33
|
Peter HH, Ochs HD, Cunningham-Rundles C, Vinh DC, Kiessling P, Greve B, Jolles S. Targeting FcRn for immunomodulation: Benefits, risks, and practical considerations. J Allergy Clin Immunol 2020; 146:479-491.e5. [PMID: 32896308 PMCID: PMC7471860 DOI: 10.1016/j.jaci.2020.07.016] [Citation(s) in RCA: 72] [Impact Index Per Article: 14.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2020] [Revised: 07/21/2020] [Accepted: 07/23/2020] [Indexed: 02/08/2023]
Abstract
The neonatal fragment crystallizable (Fc) receptor (FcRn) functions as a recycling mechanism to prevent degradation and extend the half-life of IgG and albumin in the circulation. Several FcRn inhibitors selectively targeting IgG recycling are now moving rapidly toward clinical practice in neurology and hematology. These molecules accelerate the destruction of IgG, reducing pathogenic IgG and IgG immune complexes, with no anticipated effects on IgA, IgM, IgE, complement, plasma cells, B cells, or other cells of the innate or adaptive immune systems. FcRn inhibitors have potential for future use in a much wider variety of antibody-mediated autoimmune diseases. Given the imminent clinical use, potential for broader utility, and novel mechanism of action of FcRn inhibitors, here we review data from 4 main sources: (a) currently available activity, safety, and mechanism-of-action data from clinical trials of FcRn inhibitors; (b) other procedures and treatments that also remove IgG (plasma donation, plasma exchange, immunoadsorption); (c) diseases resulting in loss of IgG; and (d) primary immunodeficiencies with potential mechanistic similarities to those induced by FcRn inhibitors. These data have been evaluated to provide practical considerations for the assessment, monitoring, and reduction of any potential infection risk associated with FcRn inhibition, in addition to highlighting areas for future research.
Collapse
Affiliation(s)
- Hans-Hartmut Peter
- Freiburg University Hospital, Centre for Chronic Immunodeficiency, Freiburg, Germany
| | - Hans D Ochs
- Seattle Children's Research Institute, Seattle, Wash; Department of Pediatrics, University of Washington, Seattle, Wash
| | | | - Donald C Vinh
- Division of Infectious Diseases, Department of Medicine and Department of Medical Microbiology, McGill University Health Centre, Montreal, Quebec, Canada; Infectious Diseases & Immunity in Global Health Program, Research Institute-McGill University Health Centre, Montreal, Quebec, Canada
| | | | | | - Stephen Jolles
- Immunodeficiency Centre for Wales, University Hospital of Wales, Cardiff, United Kingdom.
| |
Collapse
|
34
|
Freyer CW, Porter DL. Cytokine release syndrome and neurotoxicity following CAR T-cell therapy for hematologic malignancies. J Allergy Clin Immunol 2020; 146:940-948. [PMID: 32771558 DOI: 10.1016/j.jaci.2020.07.025] [Citation(s) in RCA: 105] [Impact Index Per Article: 21.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2020] [Revised: 07/08/2020] [Accepted: 07/14/2020] [Indexed: 12/18/2022]
Abstract
Chimeric antigen receptor T cells are a new and exciting immunotherapeutic approach to managing cancer, with impressive efficacy but potentially life-threatening inflammatory toxicities such as cytokine release syndrome (CRS) and immune effector cell-associated neurotoxicity syndrome (ICANS). Patients with severe CRS may develop capillary leak syndrome and disseminated intravascular coagulation, with a cytokine signature similar to that of macrophage activation syndrome/hemophagocytic lymphohistiocytosis. Moderate-to-severe CRS is managed with the IL-6 receptor antagonist tocilizumab with or without corticosteroids, with questions remaining regarding the optimal management of nonresponders. ICANS is an inflammatory neurotoxicity typically occurring after CRS and characterized by impaired blood-brain barrier integrity. Symptoms of encephalopathy range from mild confusion and aphasia to somnolence, obtundation, and in some cases seizures and cerebral edema. ICANS is currently managed with corticosteroids; however, the optimal dose and duration remain to be determined. Little information is available to guide the management of patients with steroid-refractory ICANS. Numerous cytokine-targeted therapies have been proposed to manage these inflammatory toxicities, but few clinical data are available. Management of inflammatory toxicities of chimeric antigen receptor T cells often requires multidisciplinary management and intensive care, during which allergists and immunologists may encounter patients with these unique toxicities.
Collapse
Affiliation(s)
- Craig W Freyer
- Department of Pharmacy, Hospital of the University of Pennsylvania, Philadelphia, Pa.
| | - David L Porter
- Division of Cellular Therapy and Stem Cell Transplant, Hospital of the University of Pennsylvania, Philadelphia, Pa; Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pa
| |
Collapse
|
35
|
Davis JS, Ferreira D, Paige E, Gedye C, Boyle M. Infectious Complications of Biological and Small Molecule Targeted Immunomodulatory Therapies. Clin Microbiol Rev 2020; 33:e00035-19. [PMID: 32522746 PMCID: PMC7289788 DOI: 10.1128/cmr.00035-19] [Citation(s) in RCA: 82] [Impact Index Per Article: 16.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
The past 2 decades have seen a revolution in our approach to therapeutic immunosuppression. We have moved from relying on broadly active traditional medications, such as prednisolone or methotrexate, toward more specific agents that often target a single receptor, cytokine, or cell type, using monoclonal antibodies, fusion proteins, or targeted small molecules. This change has transformed the treatment of many conditions, including rheumatoid arthritis, cancers, asthma, and inflammatory bowel disease, but along with the benefits have come risks. Contrary to the hope that these more specific agents would have minimal and predictable infectious sequelae, infectious complications have emerged as a major stumbling block for many of these agents. Furthermore, the growing number and complexity of available biologic agents makes it difficult for clinicians to maintain current knowledge, and most review articles focus on a particular target disease or class of agent. In this article, we review the current state of knowledge about infectious complications of biologic and small molecule immunomodulatory agents, aiming to create a single resource relevant to a broad range of clinicians and researchers. For each of 19 classes of agent, we discuss the mechanism of action, the risk and types of infectious complications, and recommendations for prevention of infection.
Collapse
Affiliation(s)
- Joshua S Davis
- Department of Infectious Diseases and Immunology, John Hunter Hospital, Newcastle, NSW, Australia
- Global and Tropical Health Division, Menzies School of Health Research and Charles Darwin University, Darwin, NT, Australia
- School of Medicine and Public Health, University of Newcastle, Newcastle, NSW, Australia
| | - David Ferreira
- School of Medicine, University of New South Wales, Sydney, NSW, Australia
| | - Emma Paige
- Department of Infectious Diseases, Alfred Hospital, Melbourne, VIC, Australia
| | - Craig Gedye
- School of Medicine, University of New South Wales, Sydney, NSW, Australia
- Department of Oncology, Calvary Mater Hospital, Newcastle, NSW, Australia
| | - Michael Boyle
- Department of Infectious Diseases and Immunology, John Hunter Hospital, Newcastle, NSW, Australia
- School of Medicine and Public Health, University of Newcastle, Newcastle, NSW, Australia
| |
Collapse
|
36
|
Bilich T, Nelde A, Bauer J, Walz S, Roerden M, Salih HR, Weisel K, Besemer B, Marcu A, Lübke M, Schuhmacher J, Neidert MC, Rammensee HG, Stevanović S, Walz JS. Mass spectrometry-based identification of a B-cell maturation antigen-derived T-cell epitope for antigen-specific immunotherapy of multiple myeloma. Blood Cancer J 2020; 10:24. [PMID: 32111817 PMCID: PMC7048774 DOI: 10.1038/s41408-020-0288-3] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2019] [Accepted: 10/28/2019] [Indexed: 01/10/2023] Open
Abstract
The B-cell maturation antigen (BCMA) is currently being evaluated as promising tumor-associated surface antigen for T-cell-based immunotherapy approaches, such as CAR T cells and bispecific antibodies, in multiple myeloma (MM). Cytotoxic T cells bearing BCMA-specific T-cell receptors might further allow targeting HLA-presented antigens derived from the intracellular domain of BCMA. By analyzing a mass spectrometry-acquired immunopeptidome dataset of primary MM samples and MM cell lines for BCMA-derived HLA ligands, we identified the naturally presented HLA-B*18-restricted ligand P(BCMA)B*18. Additionally, P(BCMA)B*18 was identified on primary CLL samples, thereby expanding the range for possible applications. P(BCMA)B*18 induced multifunctional BCMA-specific cells de novo from naïve CD8+ T cells of healthy volunteers. These T cells exhibited antigen-specific lysis of autologous peptide-loaded cells. Even in the immunosuppressive context of MM, we detected spontaneous memory T-cell responses against P(BCMA)B*18 in patients. By applying CTLA-4 and PD-1 inhibition in vitro we induced multifunctional P(BCMA)B*18-specific CD8+ T cells in MM patients lacking preexisting BCMA-directed immune responses. Finally, we could show antigen-specific lysis of autologous peptide-loaded target cells and even MM.1S cells naturally presenting P(BCMA)B*18 using patient-derived P(BCMA)B*18-specific T cells. Hence, this BCMA-derived T-cell epitope represents a promising target for T-cell-based immunotherapy and monitoring following immunotherapy in B-cell malignancy patients.
Collapse
Affiliation(s)
- Tatjana Bilich
- University Hospital Tübingen, Clinical Collaboration Unit Translational Immunology, German Cancer Consortium (DKTK), Tübingen, Germany
- University of Tübingen, Institute for Cell Biology, Department of Immunology, Tübingen, Germany
| | - Annika Nelde
- University Hospital Tübingen, Clinical Collaboration Unit Translational Immunology, German Cancer Consortium (DKTK), Tübingen, Germany
- University of Tübingen, Institute for Cell Biology, Department of Immunology, Tübingen, Germany
| | - Jens Bauer
- University Hospital Tübingen, Clinical Collaboration Unit Translational Immunology, German Cancer Consortium (DKTK), Tübingen, Germany
- University of Tübingen, Institute for Cell Biology, Department of Immunology, Tübingen, Germany
| | - Simon Walz
- University of Tübingen, Institute for Cell Biology, Department of Immunology, Tübingen, Germany
- University Hospital Tübingen, Department of Urology, Tübingen, Germany
| | - Malte Roerden
- University Hospital Tübingen, Department of Hematology and Oncology, Tübingen, Germany
| | - Helmut R Salih
- University Hospital Tübingen, Clinical Collaboration Unit Translational Immunology, German Cancer Consortium (DKTK), Tübingen, Germany
| | - Katja Weisel
- University Hospital Tübingen, Department of Hematology and Oncology, Tübingen, Germany
- University Hospital Hamburg-Eppendorf, Department of Oncology, Hamburg-Eppendorf, Germany
| | - Britta Besemer
- University Hospital Tübingen, Department of Hematology and Oncology, Tübingen, Germany
| | - Ana Marcu
- University of Tübingen, Institute for Cell Biology, Department of Immunology, Tübingen, Germany
| | - Maren Lübke
- University of Tübingen, Institute for Cell Biology, Department of Immunology, Tübingen, Germany
| | - Juliane Schuhmacher
- University of Tübingen, Institute for Cell Biology, Department of Immunology, Tübingen, Germany
| | - Marian C Neidert
- University Hospital Zurich and University of Zurich, Department of Neurosurgery, Clinical Neuroscience Center, Zurich, Switzerland
| | - Hans-Georg Rammensee
- University of Tübingen, Institute for Cell Biology, Department of Immunology, Tübingen, Germany
- German Cancer Consortium (DKTK), DKFZ partner site Tübingen, Tübingen, Germany
| | - Stefan Stevanović
- University of Tübingen, Institute for Cell Biology, Department of Immunology, Tübingen, Germany
- German Cancer Consortium (DKTK), DKFZ partner site Tübingen, Tübingen, Germany
| | - Juliane S Walz
- University Hospital Tübingen, Clinical Collaboration Unit Translational Immunology, German Cancer Consortium (DKTK), Tübingen, Germany.
| |
Collapse
|
37
|
Jain T, Litzow MR. Management of toxicities associated with novel immunotherapy agents in acute lymphoblastic leukemia. Ther Adv Hematol 2020; 11:2040620719899897. [PMID: 32010436 PMCID: PMC6971963 DOI: 10.1177/2040620719899897] [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: 11/05/2019] [Accepted: 12/18/2019] [Indexed: 12/15/2022] Open
Abstract
The advent of novel immunotherapies, such as blinatumomab, inotuzumab, and chimeric antigen receptor (CAR) T cell therapy, has revolutionized the therapeutic landscape in the treatment of relapsed/refractory B cell acute lymphoblastic leukemia, but can be associated with specific toxicities. We review unique toxicities of each of these in this article. Blinatumomab, a bispecific T cell engager, has been associated with cytokine release syndrome (CRS) and neurological toxicities, both of which can be prevented and managed with corticosteroids. Inotuzumab is a calicheamicin-conjugated CD22 targeting antibody. The calicheamicin component of the drug is likely associated with the hepatotoxicity seen with inotuzumab, especially sinusoidal obstruction syndrome, which can happen both in the context of the drug alone, and also with allogeneic stem cell transplantation. QT prolongation has also been noted with inotuzumab. CAR T therapy uses genetically modified autologous T cells directed against CD19, a known target on B cells. CRS and neurological symptoms, formally termed as immune-effector-cell-associated neurological syndrome, have been described along with hypogammaglobulinemia, cytopenias, and infections.
Collapse
Affiliation(s)
- Tania Jain
- Hematologic Malignancies and Bone Marrow Transplantation Program, Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, 1650 Orleans Street, 3M88, Baltimore, MD 21287, USA
| | - Mark R Litzow
- Division of Hematology and Bone Marrow Transplant, Mayo Clinic, Rochester, MN, USA
| |
Collapse
|
38
|
Wyatt KD, Bram RJ. Immunotherapy in pediatric B-cell acute lymphoblastic leukemia. Hum Immunol 2019; 80:400-408. [DOI: 10.1016/j.humimm.2019.01.011] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2018] [Revised: 01/31/2019] [Accepted: 01/31/2019] [Indexed: 12/11/2022]
|
39
|
Maschmeyer G, De Greef J, Mellinghoff SC, Nosari A, Thiebaut-Bertrand A, Bergeron A, Franquet T, Blijlevens NMA, Maertens JA. Infections associated with immunotherapeutic and molecular targeted agents in hematology and oncology. A position paper by the European Conference on Infections in Leukemia (ECIL). Leukemia 2019; 33:844-862. [PMID: 30700842 PMCID: PMC6484704 DOI: 10.1038/s41375-019-0388-x] [Citation(s) in RCA: 124] [Impact Index Per Article: 20.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2018] [Revised: 12/31/2018] [Accepted: 01/11/2019] [Indexed: 02/08/2023]
Abstract
A multitude of new agents for the treatment of hematologic malignancies has been introduced over the past decade. Hematologists, infectious disease specialists, stem cell transplant experts, pulmonologists and radiologists have met within the framework of the European Conference on Infections in Leukemia (ECIL) to provide a critical state-of-the-art on infectious complications associated with immunotherapeutic and molecular targeted agents used in clinical routine. For brentuximab vedotin, blinatumomab, CTLA4- and PD-1/PD-L1-inhibitors as well as for ibrutinib, idelalisib, HDAC inhibitors, mTOR inhibitors, ruxolitinib, and venetoclax, a detailed review of data available until August 2018 has been conducted, and specific recommendations for prophylaxis, diagnostic and differential diagnostic procedures as well as for clinical management have been developed.
Collapse
Affiliation(s)
- Georg Maschmeyer
- Department of Hematology, Oncology and Palliative Care, Klinikum Ernst von Bergmann, Charlottenstrasse 72, 14467, Potsdam, Germany.
| | - Julien De Greef
- Department of Internal Medicine and Infectious Diseases, Saint-Luc University Hospital, Université Catholique de Louvain, Brussels, Belgium
- Assistance Publique-Hôpitaux de Paris (AP-HP), Department of Hematology, Henri Mondor Teaching Hospital, Créteil, France
| | - Sibylle C Mellinghoff
- Department I of Internal Medicine, University Hospital of Cologne, Cologne, Germany
- Cologne Excellence Cluster on Cellular Stress Responses in Aging-Associated Diseases (CECAD), University of Cologne, Cologne, Germany
| | - Annamaria Nosari
- Department of Hematology, ASST Grande Ospedale Metropolitano Niguarda, Milan, Italy
| | | | - Anne Bergeron
- Department of Pneumology, Université Paris Diderot, APHP Saint-Louis Hospital, Paris, France
| | - Tomas Franquet
- Department of Radiology, Hospital de Sant Pau, Barcelona, Spain
| | | | - Johan A Maertens
- Department of Hematology, University Hospitals Leuven, Leuven, Belgium
- Department of Microbiology and Immunology, University of Leuven, Leuven, Belgium
| |
Collapse
|
40
|
Patel SY, Carbone J, Jolles S. The Expanding Field of Secondary Antibody Deficiency: Causes, Diagnosis, and Management. Front Immunol 2019; 10:33. [PMID: 30800120 PMCID: PMC6376447 DOI: 10.3389/fimmu.2019.00033] [Citation(s) in RCA: 139] [Impact Index Per Article: 23.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2018] [Accepted: 01/08/2019] [Indexed: 12/11/2022] Open
Abstract
Antibody deficiency or hypogammaglobulinemia can have primary or secondary etiologies. Primary antibody deficiency (PAD) is the result of intrinsic genetic defects, whereas secondary antibody deficiency may arise as a consequence of underlying conditions or medication use. On a global level, malnutrition, HIV, and malaria are major causes of secondary immunodeficiency. In this review we consider secondary antibody deficiency, for which common causes include hematological malignancies, such as chronic lymphocytic leukemia or multiple myeloma, and their treatment, protein-losing states, and side effects of a number of immunosuppressive agents and procedures involved in solid organ transplantation. Secondary antibody deficiency is not only much more common than PAD, but is also being increasingly recognized with the wider and more prolonged use of a growing list of agents targeting B cells. SAD may thus present to a broad range of specialties and is associated with an increased risk of infection. Early diagnosis and intervention is key to avoiding morbidity and mortality. Optimizing treatment requires careful clinical and laboratory assessment and may involve close monitoring of risk parameters, vaccination, antibiotic strategies, and in some patients, immunoglobulin replacement therapy (IgRT). This review discusses the rapidly evolving list of underlying causes of secondary antibody deficiency, specifically focusing on therapies targeting B cells, alongside recent advances in screening, biomarkers of risk for the development of secondary antibody deficiency, diagnosis, monitoring, and management.
Collapse
Affiliation(s)
- Smita Y. Patel
- Clinical Immunology Department, Oxford University Hospitals NHS Foundation Trust, Oxford, United Kingdom
| | - Javier Carbone
- Clinical Immunology Department, Hospital General Universitario Gregorio Marañon, Madrid, Spain
| | - Stephen Jolles
- Immunodeficiency Centre for Wales, University Hospital of Wales, Cardiff, United Kingdom
| |
Collapse
|
41
|
Del Bufalo F, Merli P, Alessi I, Locatelli F. B-cell depleting immunotherapies: therapeutic opportunities and toxicities. Expert Rev Clin Immunol 2019; 15:497-509. [PMID: 30681371 DOI: 10.1080/1744666x.2019.1573672] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
INTRODUCTION The last few years have witnessed what can certainly be defined as a 'period of renaissance' for immunotherapy in the field of hematological malignancies. In particular, antibody-mediated and cell-mediated immunotherapy have significantly changed the treatment approach of patients with B-cell lymphoproliferative disorders. These therapies, initially employed in patients with refractory/relapsed disease, are now integrated in the treatment of newly diagnosed patients. Together with the therapeutic success, we have also learnt that these innovative therapies can induce relevant, sometimes life-threatening or even fatal, side effects. Areas covered: In this review article, we analyzed the applicative therapeutic scenario and the peculiar toxicities associated with approaches of immunotherapy, paying particular attention to the new emerging side effects, substantially unknown before the introduction of these therapies. Expert commentary: Both monoclonal antibodies and cell therapy with lymphocytes genetically modified to be redirected against leukemia targets through the transduction with chimeric antigen receptors (CARs) have obtained unprecedented success in rescuing patients with resistant B-cell malignancies. Complications, such as neurotoxicity, cytokine release syndrome or persistent B-cell lymphopenia, must always be taken into consideration and diagnosed in a timely manner in patients with B-cell neoplasms to guarantee optimal management, thus avoiding they blunting the efficacy of immunotherapy.
Collapse
Affiliation(s)
- Francesca Del Bufalo
- a Department of Pediatric Hematology and Oncology, Cellular and Gene Therapy , IRCCS Ospedale Pediatrico Bambino Gesù , Rome , Italy
| | - Pietro Merli
- a Department of Pediatric Hematology and Oncology, Cellular and Gene Therapy , IRCCS Ospedale Pediatrico Bambino Gesù , Rome , Italy
| | - Iside Alessi
- a Department of Pediatric Hematology and Oncology, Cellular and Gene Therapy , IRCCS Ospedale Pediatrico Bambino Gesù , Rome , Italy
| | - Franco Locatelli
- a Department of Pediatric Hematology and Oncology, Cellular and Gene Therapy , IRCCS Ospedale Pediatrico Bambino Gesù , Rome , Italy.,b Department of Pediatrics , Sapienza University of Rome , Rome , Italy
| |
Collapse
|
42
|
Jain T, Litzow MR. No free rides: management of toxicities of novel immunotherapies in ALL, including financial. HEMATOLOGY. AMERICAN SOCIETY OF HEMATOLOGY. EDUCATION PROGRAM 2018; 2018:25-34. [PMID: 30504288 PMCID: PMC6246022 DOI: 10.1182/asheducation-2018.1.25] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/10/2023]
Abstract
Therapeutic options for acute lymphoblastic leukemia, especially in the relapsed/refractory setting, have expanded significantly in recent times. However, this comes at the cost of toxicities: medical as well as financial. We highlight some of the unique toxicities associated with the novel agents to apprise our readers about what to expect, how to recognize them, and how to manage these toxicities. One of the toxicities seen with inotuzumab, a CD22 antibody drug conjugate, is sinusoidal obstruction syndrome, which can be fatal in >80% of patients if associated with multiorgan failure. Blinatumomab, a monoclonal antibody targeting CD19, is associated with cytokine release syndrome (CRS) and neurotoxicity, both of which require prompt recognition and management primarily with corticosteroids. CRS and neurotoxicity are more common and more severe with chimeric antigen receptor T-cell therapy (CAR-T). The fact that CAR-T cannot be discontinued on demand adds a layer of complexity to the management of related toxicities of this therapy. Tocilizumab, an interleukin-6 receptor blocker, is used to treat severe CRS from CAR-T, whereas corticosteroids remain the mainstay for neurotoxicity management. Although effective, these drugs carry a high price tag, and we review the available data on cost-effectiveness of these agents, keeping in mind that median follow-up on most of these studies is limited and that long-term data on durability of response remain to be seen.
Collapse
MESH Headings
- Antibodies, Bispecific/economics
- Antibodies, Bispecific/therapeutic use
- Antibodies, Monoclonal, Humanized/adverse effects
- Antibodies, Monoclonal, Humanized/economics
- Antibodies, Monoclonal, Humanized/therapeutic use
- Costs and Cost Analysis
- Hepatic Veno-Occlusive Disease/chemically induced
- Hepatic Veno-Occlusive Disease/economics
- Hepatic Veno-Occlusive Disease/pathology
- Humans
- Immunotherapy, Adoptive/adverse effects
- Immunotherapy, Adoptive/economics
- Immunotherapy, Adoptive/methods
- Inotuzumab Ozogamicin
- Neurotoxicity Syndromes/economics
- Neurotoxicity Syndromes/immunology
- Neurotoxicity Syndromes/pathology
- Precursor Cell Lymphoblastic Leukemia-Lymphoma/economics
- Precursor Cell Lymphoblastic Leukemia-Lymphoma/pathology
- Precursor Cell Lymphoblastic Leukemia-Lymphoma/therapy
- Receptors, Chimeric Antigen
Collapse
Affiliation(s)
- Tania Jain
- Adult Bone Marrow Transplantation Service, Memorial Sloan-Kettering Cancer Center, New York, NY; and
| | - Mark R. Litzow
- Division of Hematology and Bone Marrow Transplant, Mayo Clinic, Rochester, MN
| |
Collapse
|
43
|
Jain T, Litzow MR. No free rides: management of toxicities of novel immunotherapies in ALL, including financial. Blood Adv 2018; 2:3393-3403. [PMID: 30482769 PMCID: PMC6258912 DOI: 10.1182/bloodadvances.2018020198] [Citation(s) in RCA: 40] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2018] [Accepted: 10/13/2018] [Indexed: 12/27/2022] Open
Abstract
Therapeutic options for acute lymphoblastic leukemia, especially in the relapsed/refractory setting, have expanded significantly in recent times. However, this comes at the cost of toxicities: medical as well as financial. We highlight some of the unique toxicities associated with the novel agents to apprise our readers about what to expect, how to recognize them, and how to manage these toxicities. One of the toxicities seen with inotuzumab, a CD22 antibody drug conjugate, is sinusoidal obstruction syndrome, which can be fatal in >80% of patients if associated with multiorgan failure. Blinatumomab, a monoclonal antibody targeting CD19, is associated with cytokine release syndrome (CRS) and neurotoxicity, both of which require prompt recognition and management primarily with corticosteroids. CRS and neurotoxicity are more common and more severe with chimeric antigen receptor T-cell therapy (CAR-T). The fact that CAR-T cannot be discontinued on demand adds a layer of complexity to the management of related toxicities of this therapy. Tocilizumab, an interleukin-6 receptor blocker, is used to treat severe CRS from CAR-T, whereas corticosteroids remain the mainstay for neurotoxicity management. Although effective, these drugs carry a high price tag, and we review the available data on cost-effectiveness of these agents, keeping in mind that median follow-up on most of these studies is limited and that long-term data on durability of response remain to be seen.
Collapse
Affiliation(s)
- Tania Jain
- Adult Bone Marrow Transplantation Service, Memorial Sloan Kettering Cancer Center, New York, NY; and
| | - Mark R Litzow
- Division of Hematology and Bone Marrow Transplant, Mayo Clinic, Rochester, MN
| |
Collapse
|
44
|
Algeri M, Del Bufalo F, Galaverna F, Locatelli F. Current and future role of bispecific T-cell engagers in pediatric acute lymphoblastic leukemia. Expert Rev Hematol 2018; 11:945-956. [PMID: 30358451 DOI: 10.1080/17474086.2018.1540928] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
Abstract
INTRODUCTION The clinical application of immunotherapy has resulted into a significant improvement in the outcome of children with relapsed/refractory B-cell precursor acute lymphoblastic leukemia (r/r BCP-ALL). In this setting, the use of bispecific T-cell-engager antibodies (BiTEs), such as blinatumomab, which harness the cytotoxic activity of T cells against CD19-positive lymphoblasts, has emerged as a most promising and impactful strategy. Areas covered: This review discusses the main structural and functional features of BiTEs, as well as the current status of their clinical application in childhood ALL. Moreover, future prospects to increase the efficacy of BiTEs are addressed. Expert commentary: The promising results obtained in patients with advanced BCP-ALL pave the way for further improvement in the context of less resistant/advanced disease. Future research is rapidly progressing on several aspects, including the use of blinatumomab in first-line protocols, identification of factors predicting response, use of combinatorial approaches and bioengineering of new molecules with dual specificity or increased potency, stability and half-life. The results of these studies, expected to be available in the next future, will provide further advancement in the development of effective, impactful, targeted immunotherapy for treatment of childhood BCP-ALL, with the concrete potential to revolutionize the clinical practice.
Collapse
Affiliation(s)
- Mattia Algeri
- a Department of Pediatric Hematology and Oncology , IRCCS, Ospedale Pediatrico Bambino Gesù , Rome , Italy
| | - Francesca Del Bufalo
- a Department of Pediatric Hematology and Oncology , IRCCS, Ospedale Pediatrico Bambino Gesù , Rome , Italy
| | - Federica Galaverna
- a Department of Pediatric Hematology and Oncology , IRCCS, Ospedale Pediatrico Bambino Gesù , Rome , Italy
| | - Franco Locatelli
- a Department of Pediatric Hematology and Oncology , IRCCS, Ospedale Pediatrico Bambino Gesù , Rome , Italy.,b Department of Pediatrics , University of Pavia , Pavia , Italy
| |
Collapse
|
45
|
Liu D, Zhao J. Cytokine release syndrome: grading, modeling, and new therapy. J Hematol Oncol 2018; 11:121. [PMID: 30249264 PMCID: PMC6154787 DOI: 10.1186/s13045-018-0653-x] [Citation(s) in RCA: 98] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2018] [Accepted: 08/15/2018] [Indexed: 12/19/2022] Open
Abstract
Genetically modified T cells that express a chimeric antigen receptor (CAR) are opening a new frontier in cancer immunotherapy. CAR T cells currently are in clinical trials for many cancer types. Cytokine release syndrome (CRS) and neurotoxicities (CAR-related encephalopathy syndrome, CRES) are major adverse events limiting wide deployment of the CAR T cell treatment. Major efforts are ongoing to characterize the pathogenesis and etiology of CRS and CRES. Mouse models have been established to facilitate the study of pathogenesis of the major toxicities of CAR T cells. Myeloid cells including macrophages and monocytes, not the CAR T cells, were found to be the major cells mediating CRS and CRES by releasing IL-1 and IL-6 among other cytokines. Blocking IL-1 or depletion of monocytes abolished both CRS and CRES, whereas IL-6 blocker can ameliorate CRS but not CRES. Therefore, both IL-1 and IL-6 are major cytokines for CRS, though IL-1 is responsible for CRES. It was also demonstrated in the mouse models that blocking CRS does not interfere with the CAR T cell antitumor functions. We summarized new developments in the grading, modeling, and possible new therapeutic approaches for CRS and CRES in this review.
Collapse
Affiliation(s)
- Delong Liu
- Department of Oncology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, China.
| | - Juanjuan Zhao
- The Affiliated Cancer Hospital of Zhengzhou University and Henan Cancer Hospital, 127 Dongming Road, Zhengzhou, 450008, China
| |
Collapse
|
46
|
Abstract
Acute lymphoblastic leukemia (ALL) is the most common cancer in childhood. Standard chemotherapy has afforded outstanding outcomes for many patients; however, there remain some sub-groups with high-risk features, refractory disease, and patients that relapse who have a poor prognosis with conventional treatments. Over the past decade, there have been significant advances in newer treatment options, including improved monoclonal antibody therapies, T cell engagers, and chimeric antigen T-cell receptor products, all of which have changed the landscape for patients who relapse. These are now being introduced more frequently and at earlier stages of therapy. We present a brief overview of the biology and etiology of childhood ALL, treatment strategies currently in use, and discuss some newer strategies and their possible role in the future of ALL therapy for children.
Collapse
Affiliation(s)
- Kelly W Maloney
- Center for Cancer and Blood Disorders, Children's Hospital Colorado, Pediatric Hematology/Oncology/Bone Marrow Transplant, University of Colorado School of Medicine, 13123 East 16th Av, Box B115, Aurora, CO, 80045, USA
| | - Lia Gore
- Center for Cancer and Blood Disorders, Children's Hospital Colorado, Pediatric Hematology/Oncology/Bone Marrow Transplant, University of Colorado School of Medicine, 13123 East 16th Av, Box B115, Aurora, CO, 80045, USA.
| |
Collapse
|
47
|
ESCMID Study Group for Infections in Compromised Hosts (ESGICH) Consensus Document on the safety of targeted and biological therapies: an infectious diseases perspective (Agents targeting lymphoid cells surface antigens [I]: CD19, CD20 and CD52). Clin Microbiol Infect 2018; 24 Suppl 2:S71-S82. [PMID: 29447988 DOI: 10.1016/j.cmi.2018.02.003] [Citation(s) in RCA: 148] [Impact Index Per Article: 21.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2017] [Revised: 01/28/2018] [Accepted: 02/03/2018] [Indexed: 12/13/2022]
Abstract
BACKGROUND The present review is part of the ESCMID Study Group for Infections in Compromised Hosts (ESGICH) Consensus Document on the safety of targeted and biological therapies. AIMS To review, from an Infectious Diseases perspective, the safety profile of agents targeting CD19, CD20 and CD52 and to suggest preventive recommendations. SOURCES Computer-based MEDLINE searches with MeSH terms pertaining to each agent or therapeutic family. CONTENT Although CD19-targeted agents (blinatumomab or inebilizumab) are not associated with an increased risk of infection, they may cause IgG hypogammaglobulinaemia and neutropenia. The requirement for prolonged intravenous infusion of blinatumomab may increase the risk of catheter-associated bloodstream infections. Infection remains the most common non-haematological adverse effect of anti-CD20 monoclonal antibodies, including severe respiratory tract infection, hepatitis B virus (HBV) reactivation and varicella-zoster virus infection. Screening for chronic or resolved HBV infection is recommended for patients receiving anti-CD20 monoclonal antibodies. Antiviral prophylaxis should be offered for 12-18 months to hepatitis B surface antigen (HBsAg)-positive and HBsAg-negative/anti-hepatitis B core antibody (HBc)-positive patients. Anti-Pneumocystis prophylaxis should be considered in patients receiving concomitant chemotherapy, particularly steroids. Alemtuzumab (anti-CD52) increases the risk of infections, in particular among leukaemia and solid organ transplant patients. These populations benefit from anti-Pneumocystis prophylaxis, prevention strategies for cytomegalovirus infection, and screening for HBV, hepatitis C virus and tuberculosis. Antiviral prophylaxis for at least 6-12 months should be provided for HBsAg-positive patients. IMPLICATIONS As there are limited clinical data for many of the reviewed agents, special attention must be given to promptly detect and report emerging infectious complications.
Collapse
|
48
|
Ribera JM. Efficacy and safety of bispecific T-cell engager blinatumomab and the potential to improve leukemia-free survival in B-cell acute lymphoblastic leukemia. Expert Rev Hematol 2017; 10:1057-1067. [PMID: 29082835 DOI: 10.1080/17474086.2017.1396890] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023]
Abstract
INTRODUCTION Immunotherapy is a promising modality of treatment of neoplastic diseases, including acute lymphoblastic leukemia (ALL). The CD19/CD3-bispecific T cell-engaging (BiTE®) monoclonal antibody blinatumomab can transiently bind cytotoxic T cells to CD19+ target B cells of ALL inducing their serial lysis. Areas covered: This review focuses on the efficacy and safety of blinatumomab used for the treatment of relapsed/refractory (R/R) ALL and minimal residual disease (MRD)-positive B-cell precursor (BCP) ALL in adults and children, as well as the future prospects of this drug in the treatment of ALL. Expert commentary: Blinatumomab has demonstrated encouraging response rates in MRD-positive and R/R in adults with Philadelphia chromosome-positive and -negative ALL, as well as in children with R/R ALL. Blinatumomab has a favorable safety profile, although reversible CNS events and cytokine release syndrome can occur. Ongoing trials in ALL incorporate blinatumomab in the first line therapy of BCP ALL in combination with chemotherapy, targeted therapies or other immunotherapies with the aim of increasing the depth of the remission and decreasing the probability of relapse.
Collapse
Affiliation(s)
- Josep-Maria Ribera
- a Clinical Hematology Department, ICO-Hospital Germans Trias i Pujol, Jose Carreras Research Institute, Badalona , Universitat Autonoma de Barcelona , Spain
| |
Collapse
|
49
|
Gökbuget N, Zugmaier G, Klinger M, Kufer P, Stelljes M, Viardot A, Horst HA, Neumann S, Brüggemann M, Ottmann OG, Burmeister T, Wessiepe D, Topp MS, Bargou R. Long-term relapse-free survival in a phase 2 study of blinatumomab for the treatment of patients with minimal residual disease in B-lineage acute lymphoblastic leukemia. Haematologica 2017; 102:e132-e135. [PMID: 28082340 DOI: 10.3324/haematol.2016.153957] [Citation(s) in RCA: 70] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022] Open
Affiliation(s)
- Nicola Gökbuget
- Center of Internal Medicine, Goethe University, Frankfurt, Germany
| | | | | | | | | | - Andreas Viardot
- Department for Internal Medicine III, University Ulm, Germany
| | - Heinz A Horst
- Medical Department II, Christian-Albrechts University, Kiel, Germany
| | - Svenja Neumann
- Medical Department II, Christian-Albrechts University, Kiel, Germany
| | - Monika Brüggemann
- Medical Department II, Christian-Albrechts University, Kiel, Germany
| | | | - Thomas Burmeister
- Department for Hematology and Oncology and Tumor Immunology, Charité-Universitätsmedizin Berlin, Campus Benjamin Franklin, Berlin, Germany
| | | | - Max S Topp
- Department of Internal Medicine II, Universitätsklinikum, Würzburg, Germany
| | - Ralf Bargou
- Comprehensive Cancer Center Mainfranken, Universitätsklinikum Würzburg, Germany
| |
Collapse
|
50
|
Redirecting T cells to eradicate B-cell acute lymphoblastic leukemia: bispecific T-cell engagers and chimeric antigen receptors. Leukemia 2016; 31:777-787. [PMID: 28028314 DOI: 10.1038/leu.2016.391] [Citation(s) in RCA: 51] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2016] [Revised: 11/30/2016] [Accepted: 12/05/2016] [Indexed: 12/15/2022]
Abstract
Recent advances in antibody technology to harness T cells for cancer immunotherapy, particularly in the difficult-to-treat setting of relapsed/refractory acute lymphoblastic leukemia (r/r ALL), have led to innovative methods for directing cytotoxic T cells to specific surface antigens on cancer cells. One approach involves administration of soluble bispecific (or dual-affinity) antibody-based constructs that temporarily bridge T cells and cancer cells. Another approach infuses ex vivo-engineered T cells that express a surface plasma membrane-inserted antibody construct called a chimeric antigen receptor (CAR). Both bispecific antibodies and CARs circumvent natural target cell recognition by creating a physical connection between cytotoxic T cells and target cancer cells to activate a cytolysis signaling pathway; this connection allows essentially all cytotoxic T cells in a patient to be engaged because typical tumor cell resistance mechanisms (such as T-cell receptor specificity, antigen processing and presentation, and major histocompatibility complex context) are bypassed. Both the bispecific T-cell engager (BiTE) antibody construct blinatumomab and CD19-CARs are immunotherapies that have yielded encouraging remission rates in CD19-positive r/r ALL, suggesting that they might serve as definitive treatments or bridging therapies to allogeneic hematopoietic cell transplantation. With the introduction of these immunotherapies, new challenges arise related to unique toxicities and distinctive pathways of resistance. An increasing body of knowledge is being accumulated on how to predict, prevent, and manage such toxicities, which will help to better stratify patient risk and tailor treatments to minimize severe adverse events. A deeper understanding of the precise mechanisms of action and immune resistance, interaction with other novel agents in potential combinations, and optimization in the manufacturing process will help to advance immunotherapy outcomes in the r/r ALL setting.
Collapse
|