1
|
Csizmar CM, Litzow MR, Saliba AN. Antibody-Based and Other Novel Agents in Adult B-Cell Acute Lymphoblastic Leukemia. Cancers (Basel) 2025; 17:779. [PMID: 40075627 PMCID: PMC11899621 DOI: 10.3390/cancers17050779] [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: 12/28/2024] [Revised: 02/21/2025] [Accepted: 02/22/2025] [Indexed: 03/14/2025] Open
Abstract
Despite notable progress in managing B-cell acute lymphoblastic leukemia (B-ALL) over recent decades, particularly in pediatric cohorts where the 5-year overall survival (OS) reaches 90%, outcomes for the 10-15% with relapsed and refractory disease remain unfavorable. This disparity is further accentuated in adults, where individuals over the age of 40 years undergoing aggressive multiagent chemotherapy continue to have lower survival rates. While the adoption of pediatric-inspired treatment protocols has enhanced complete remission (CR) rates among younger adults, 20-30% of these patients experience relapse, resulting in a subsequent 5-year OS rate of 40-50%. For relapsed B-ALL in adults, there is no universally accepted standard salvage therapy, and the median OS is short. The cornerstone of B-ALL treatment continues to be the utilization of combined cytotoxic chemotherapy regimens to maximize early and durable disease control. In this manuscript, we go beyond the multiagent chemotherapy medications developed prior to the 1980s and focus on the incorporation of antibody-based therapy for B-ALL with an eye on existing and upcoming approved indications for blinatumomab, inotuzumab ozogamicin, other monoclonal antibodies, and chimeric antigen receptor (CAR) T cell products in frontline and relapsed/refractory settings. In addition, we discuss emerging investigational therapies that harness the therapeutic vulnerabilities of the disease through targeting apoptosis, modifying epigenetics, and inhibiting the mTOR pathway.
Collapse
Affiliation(s)
- Clifford M. Csizmar
- Division of Hematology, Department of Medicine, Mayo Clinic, Rochester, MN 55905, USA;
| | | | - Antoine N. Saliba
- Division of Hematology, Department of Medicine, Mayo Clinic, Rochester, MN 55905, USA;
| |
Collapse
|
2
|
Erkner E, Hentrich T, Schairer R, Fitzel R, Secker-Grob KA, Jeong J, Keppeler H, Korkmaz F, Schulze-Hentrich JM, Lengerke C, Schneidawind D, Schneidawind C. The RORɣ/SREBP2 pathway is a master regulator of cholesterol metabolism and serves as potential therapeutic target in t(4;11) leukemia. Oncogene 2024; 43:281-293. [PMID: 38030791 PMCID: PMC10798886 DOI: 10.1038/s41388-023-02903-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2023] [Revised: 11/10/2023] [Accepted: 11/16/2023] [Indexed: 12/01/2023]
Abstract
Dysregulated cholesterol homeostasis promotes tumorigenesis and progression. Therefore, metabolic reprogramming constitutes a new hallmark of cancer. However, until today, only few therapeutic approaches exist to target this pathway due to the often-observed negative feedback induced by agents like statins leading to controversially increased cholesterol synthesis upon inhibition. Sterol regulatory element-binding proteins (SREBPs) are key transcription factors regulating the synthesis of cholesterol and fatty acids. Since SREBP2 is difficult to target, we performed pharmacological inhibition of retinoic acid receptor (RAR)-related orphan receptor gamma (RORγ), which acts upstream of SREBP2 and serves as master regulator of the cholesterol metabolism. This resulted in an inactivated cholesterol-related gene program with significant downregulation of cholesterol biosynthesis. Strikingly, these effects were more pronounced than the effects of fatostatin, a direct SREBP2 inhibitor. Upon RORγ inhibition, RNA sequencing showed strongly increased cholesterol efflux genes leading to leukemic cell death and cell cycle changes in a dose- and time-dependent manner. Combinatorial treatment of t(4;11) cells with the RORγ inhibitor showed additive effects with cytarabine and even strong anti-leukemia synergism with atorvastatin by circumventing the statin-induced feedback. Our results suggest a novel therapeutic strategy to inhibit tumor-specific cholesterol metabolism for the treatment of t(4;11) leukemia.
Collapse
Affiliation(s)
- Estelle Erkner
- Department of Hematology, Oncology, Clinical Immunology and Rheumatology, University Hospital Tuebingen, Tuebingen, Germany
| | - Thomas Hentrich
- Department of Genetics/Epigenetics, Faculty NT, Saarland University, Saarbruecken, Germany
| | - Rebekka Schairer
- Department of Hematology, Oncology, Clinical Immunology and Rheumatology, University Hospital Tuebingen, Tuebingen, Germany
| | - Rahel Fitzel
- Department of Hematology, Oncology, Clinical Immunology and Rheumatology, University Hospital Tuebingen, Tuebingen, Germany
| | - Kathy-Ann Secker-Grob
- Department of Hematology, Oncology, Clinical Immunology and Rheumatology, University Hospital Tuebingen, Tuebingen, Germany
| | - Johan Jeong
- Process Cell Sciences, Merck & Co., Inc., Kenilworth, NJ, USA
| | - Hildegard Keppeler
- Department of Hematology, Oncology, Clinical Immunology and Rheumatology, University Hospital Tuebingen, Tuebingen, Germany
| | - Fulya Korkmaz
- Department of Hematology, Oncology, Clinical Immunology and Rheumatology, University Hospital Tuebingen, Tuebingen, Germany
| | | | - Claudia Lengerke
- Department of Hematology, Oncology, Clinical Immunology and Rheumatology, University Hospital Tuebingen, Tuebingen, Germany
| | - Dominik Schneidawind
- Department of Hematology, Oncology, Clinical Immunology and Rheumatology, University Hospital Tuebingen, Tuebingen, Germany
- Department of Medical Oncology and Hematology, University Hospital Zurich, Zurich, Switzerland
| | - Corina Schneidawind
- Department of Hematology, Oncology, Clinical Immunology and Rheumatology, University Hospital Tuebingen, Tuebingen, Germany.
- Department of Medical Oncology and Hematology, University Hospital Zurich, Zurich, Switzerland.
| |
Collapse
|
3
|
Andrade AM, Teixeira VR, Pogue R, Figueiredo ACMG, Carvalho JL. A systematic review on the cost-effectiveness assessment of tisagenlecleucel for refractory or relapsing B-cell acute lymphoblastic leukemia (R/R B-ALL) treatment in children and young adults. Cytotherapy 2023:S1465-3249(23)00957-X. [PMID: 37341664 DOI: 10.1016/j.jcyt.2023.05.011] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2022] [Revised: 05/02/2023] [Accepted: 05/26/2023] [Indexed: 06/22/2023]
Abstract
BACKGROUND AIMS The advanced therapy product tisagenlecleucel is a CD19-directed genetically modified autologous T-cell immunotherapy that has brought hope for children and young adults with relapsed/refractory (R/R) B-cell acute lymphoblastic leukemia (B-ALL). We sought to evaluate the cost-effectiveness of tisagenlecleucel compared with conventional salvage therapies in pediatric and young adult patients with R/R B-ALL. METHODS This systematic review followed the Preferred Reporting Items for Systematic Reviews and Meta-Analyses parameters as registered in International Prospective Register of Systematic Reviews (CRD42021266998). Literature was searched using the MEDLINE databases via PubMed, EMBASE, Lilacs, the Cochrane Central Register of Controlled Trials and Web of Science in January 2022. Titles were screened independently by two reviewers. Articles deemed to meet the inclusion criteria were screened independently on abstract, and full texts were reviewed. RESULTS In total, 5627 publications were identified, from which six eligible studies were selected. The conventional therapies identified were blinatumomab (Blina), clofarabine monotherapy (Clo-M), clofarabine combined with cyclophosphamide and etoposide (Clo-C) and the combination of fludarabine, cytarabine and idarubicin (FLA-IDA). The discounted incremental cost-effectiveness ratio (ICER) per quality-adjusted life year (QALY) gained for tisagenlecleucel compared with Clo-C and Blina averages was $38 837 and $25 569, respectively. In relation to the cost of the drug, the average of tisagenlecleucel was approximately 4.3 times, 10.8 times or 4.7 times greater than the Clo-M, Clo-C and Blina, respectively. CONCLUSIONS This systematic review highlighted that tisagenlecleucel is a much more expensive therapy than conventional alternatives. However, tisagenlecleucel performed well on the ICER, not exceeding $100 000/QALY. It was also found that the advanced therapy product was more effective than the conventional small molecule and biological drugs, in terms of life years and QALY gained.
Collapse
Affiliation(s)
- Aurélio Matos Andrade
- Medical Sciences Program, University of Brasilia, Brasilia, Distrito Federal, Brazil; Program of Evidence for Health Policy and Technologies, Oswaldo Cruz Brasilia Foundation, Brasilia, Distrito Federal, Brazil; Interdisciplinary Biosciences Laboratory, Faculty of Medicine, University of Brasília, Brasília, Distrito Federal, Brazil
| | | | - Robert Pogue
- Genomic Sciences and Biotechnology Program, Catholic University of Brasília, Brasília, Brazil
| | | | - Juliana Lott Carvalho
- Medical Sciences Program, University of Brasilia, Brasilia, Distrito Federal, Brazil; Interdisciplinary Biosciences Laboratory, Faculty of Medicine, University of Brasília, Brasília, Distrito Federal, Brazil; Faculty of Medicine, University of Brasilia, Brasilia, Distrito Federal, Brazil.
| |
Collapse
|
4
|
Chen X, Gao Q, Roshal M, Cherian S. Flow cytometric assessment for minimal/measurable residual disease in B lymphoblastic leukemia/lymphoma in the era of immunotherapy. CYTOMETRY. PART B, CLINICAL CYTOMETRY 2023; 104:205-223. [PMID: 36683279 DOI: 10.1002/cyto.b.22113] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/11/2022] [Revised: 11/30/2022] [Accepted: 12/28/2022] [Indexed: 01/24/2023]
Abstract
Minimal/measurable residual disease (MRD) is the most important independent prognostic factor for patients with B-lymphoblastic leukemia (B-LL). MRD post therapy has been incorporated into risk stratification and clinical management, resulting in substantially improved outcomes in pediatric and adult patients. Currently, MRD in B-ALL is most commonly assessed by multiparametric flow cytometry and molecular (polymerase chain reaction or high-throughput sequencing based) methods. The detection of MRD by flow cytometry in B-ALL often begins with B cell antigen-based gating strategies. Over the past several years, targeted immunotherapy directed against B cell markers has been introduced in patients with relapsed or refractory B-ALL and has demonstrated encouraging results. However, targeted therapies have significant impact on the immunophenotype of leukemic blasts, in particular, downregulation or loss of targeted antigens on blasts and normal B cell precursors, posing challenges for MRD detection using standard gating strategies. Novel flow cytometric approaches, using alternative strategies for population identification, sometimes including alternative gating reagents, have been developed and implemented to monitor MRD in the setting of post targeted therapy.
Collapse
Affiliation(s)
- Xueyan Chen
- Department of Laboratory Medicine and Pathology, University of Washington, Seattle, Washington, USA
| | - Qi Gao
- Hematopathology Service, Department of Pathology and Laboratory Medicine, Memorial Sloan Kettering Cancer Center, New York, New York, USA
| | - Mikhail Roshal
- Hematopathology Service, Department of Pathology and Laboratory Medicine, Memorial Sloan Kettering Cancer Center, New York, New York, USA
| | - Sindhu Cherian
- Department of Laboratory Medicine and Pathology, University of Washington, Seattle, Washington, USA
| |
Collapse
|
5
|
Künz T, Hauswirth AW, Hetzenauer G, Rudzki J, Nachbaur D, Steiner N. Changing Landscape in the Treatment of Adult Acute Lymphoblastic Leukemia (ALL). Cancers (Basel) 2022; 14:4290. [PMID: 36077822 PMCID: PMC9454969 DOI: 10.3390/cancers14174290] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2022] [Revised: 08/28/2022] [Accepted: 08/29/2022] [Indexed: 11/22/2022] Open
Abstract
Acute lymphoblastic leukemia (ALL) is a rare hematological malignancy characterized by proliferation and accumulation of premature lymphoid blasts. Depending on risk factors, the survival of acute lymphoblastic leukemia has significantly improved over the last decades. During the last years, measurable residual disease (MRD) assessment has evolved into one of the most sensitive markers for prognosis and risk of relapse. For this reason, measurable residual disease detection and monitoring count as standard evaluation in patients with acute lymphoblastic leukemia. Allogeneic stem cell transplantation is still the recommended treatment option for patients with high and highest risk profiles as well as for relapsed or refractory settings. The increased understanding of the pathomechanism and heterogeneity of acute lymphoblastic leukemia has led to the development of several novel therapeutic opportunities such as tyrosine-kinase inhibitors, antibody-based therapies and CAR-T cells with the aim of improving clinical outcomes. Furthermore, the major advances in disease understanding of ALL have led to the identification of different subgroups and better disease stratification. Even though novel therapy targets are constantly developed, acute lymphoblastic leukemia remains a challenging and life-threatening disease. To improve the historically unsatisfying result in therapy of adult acute lymphoblastic leukemia many clinical trials have recently been initiated to determine the optimum combination regimens of novel and old agents for adult acute lymphoblastic leukemia.
Collapse
Affiliation(s)
- Tina Künz
- Department of Internal Medicine V (Hematology and Medical Oncology), Medical University of Innsbruck, A-6020 Innsbruck, Austria
| | - Alexander W. Hauswirth
- Department of Medicine I, Division of Hematology and Hemostaseology, Medical University of Vienna, 1090 Vienna, Austria
| | - Gabriele Hetzenauer
- Department of Internal Medicine V (Hematology and Medical Oncology), Medical University of Innsbruck, A-6020 Innsbruck, Austria
| | - Jakob Rudzki
- Department of Internal Medicine V (Hematology and Medical Oncology), Medical University of Innsbruck, A-6020 Innsbruck, Austria
| | - David Nachbaur
- Department of Internal Medicine V (Hematology and Medical Oncology), Medical University of Innsbruck, A-6020 Innsbruck, Austria
| | - Normann Steiner
- Department of Internal Medicine V (Hematology and Medical Oncology), Medical University of Innsbruck, A-6020 Innsbruck, Austria
| |
Collapse
|
6
|
Pourmontaseri H, Habibzadeh N, Entezari S, Samadian F, Kiyani S, Taheri M, Ahmadi A, Fallahi MS, Sheikhzadeh F, Ansari A, Tamimi A, Deravi N. Monoclonal antibodies for the treatment of acute lymphocytic leukemia: A literature review. Hum Antibodies 2022; 30:117-130. [PMID: 35662114 DOI: 10.3233/hab-211511] [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: 11/15/2022]
Abstract
BACKGROUND Acute lymphocytic leukemia (ALL) is a type of blood cancer that is more prevalent in children. Several treatment methods are available for ALL, including chemotherapy, upfront treatment regimens, and pediatric-inspired regimens for adults. Monoclonal antibodies (Mabs) are the novel Food and Drug Administration (FDA) approved remedies for the relapsed/refractory (R/R) adult ALL. In this article, we aimed to review studies that investigated the efficacy and safety of Mabs on ALL. METHODS We gathered studies through a complete search with all proper related keywords in ISI Web of Science, SID, Scopus, Google Scholar, Science Direct, and PubMed for English language publications up to 2020. RESULTS The most commonly studied Mabs for ALL therapies are CD-19, CD-20, CD-22, and CD-52. The best results have been reported in the administration of blinatumomab, rituximab, ofatumumab, and inotuzumab with acceptable low side effects. CONCLUSION Appling personalized approach for achieving higher efficacy is one of the most important aspects of treatment. Moreover, we recommend that the wide use of these Mabs depends on designing further cost-effectiveness trials in this field.
Collapse
Affiliation(s)
- Hossein Pourmontaseri
- Student Research Committee, Fasa University of Medical Sciences, Fasa, Iran.,Bitab knowledge enterprise, Fasa University of Medical Sciences, Fasa, Iran
| | - Niloofar Habibzadeh
- Student Research Committee, School of Medical Sciences, Shahid Sadoughi University of Medical Sciences, Yazd, Iran
| | - Sarina Entezari
- Student Research Committee, School of Allied Medical Sciences, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Fatemeh Samadian
- Nursing Department, Shahid Beheshti University of Medical science, Tehran, Iran
| | - Shamim Kiyani
- Midwifery Department, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
| | - Mina Taheri
- Student Research Committee, School of Pharmacy Sciences, Shahid Sadoughi University of Medical Sciences, Yazd, Iran
| | - Ali Ahmadi
- Faculty of Biological Sciences and Technologies, Islamic Azad University Sari Branch, Sari, Iran
| | | | - Farzad Sheikhzadeh
- Student Research Committee, School of Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Arina Ansari
- Student Research Committee, School of Medicine, North Khorasan University of Medical Sciences, Bojnurd, Iran
| | - Amirhossein Tamimi
- Student Research Committee, School of Medicine, Guilan University of Medical Sciences, Rasht, Iran
| | - Niloofar Deravi
- Student Research Committee, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| |
Collapse
|
7
|
Abuasab T, Rowe J, Tvito A. Emerging Monoclonal Antibody Therapy for the Treatment of Acute Lymphoblastic Leukemia. Biologics 2021; 15:419-431. [PMID: 34703207 PMCID: PMC8536880 DOI: 10.2147/btt.s290294] [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: 05/16/2021] [Accepted: 08/03/2021] [Indexed: 12/21/2022]
Abstract
The treatment of adults with ALL has undergone tremendous progress over the past 15 years. The advances have been particularly marked with B-lineage ALL. The development of bispecific antibodies directed against CD19 ushered in a new era in overcoming persistent minimal disease in newly diagnosed ALL patients as well as successfully treating those with relapsed disease. The immune-conjugates targeting CD22 have also had a similarly impressive role in improving the outcome in such patients. These advances are now being extended to frontline regimens for B-lineage ALL, including the Philadelphia-chromosome-positive subtype. Over the past decade, the development of chimeric antigen receptor T-cell therapy (CAR-T) has ushered in a new era, opening up hope when none was available for patients with particularly advanced disease. Such advances come at a considerable price for toxicity, which, however, are lessening with experience and the development of new agents to ameliorate some of the toxicities. Unfortunately, the progress for T-cell in ALL has lagged behind that of B-lineage ALL. Of late, however, there are preliminary results of potentially exciting data using monoclonal antibodies against CD38, in the form of daratumumab, and it is hoped that these will lead to an equally successful advance in the treatment of T-ALL. Despite all these advances, ALL in adults remains a formidable disease. While ongoing progress is being made, also in the therapy of older patients, we are still lagging behind the almost totally curative potential of current therapy for childhood ALL.
Collapse
Affiliation(s)
- Tareq Abuasab
- Department of Hematology, Shaare Zedek Medical Center, Jerusalem, Israel
| | - Jacob Rowe
- Department of Hematology, Shaare Zedek Medical Center, Jerusalem, Israel
- Department of Hematology and Bone Marrow Transplantation, Rambam Health Care Campus, Haifa, Israel
- Bruce Rappaport Faculty of Medicine, Technion, Israel Institute of Technology, Haifa, Israel
| | - Ariella Tvito
- Department of Hematology, Shaare Zedek Medical Center, Jerusalem, Israel
| |
Collapse
|
8
|
Salvaris R, Fedele PL. Targeted Therapy in Acute Lymphoblastic Leukaemia. J Pers Med 2021; 11:715. [PMID: 34442359 PMCID: PMC8398498 DOI: 10.3390/jpm11080715] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2021] [Accepted: 07/21/2021] [Indexed: 11/26/2022] Open
Abstract
The last decade has seen a significant leap in our understanding of the wide range of genetic lesions underpinning acute lymphoblastic leukaemia (ALL). Next generation sequencing has led to the identification of driver mutations with significant implications on prognosis and has defined entities such as BCR-ABL-like ALL, where targeted therapies such as tyrosine kinase inhibitors (TKIs) and JAK inhibitors may play a role in its treatment. In Philadelphia positive ALL, the introduction of TKIs into frontline treatment regimens has already transformed patient outcomes. In B-ALL, agents targeting surface receptors CD19, CD20 and CD22, including monoclonal antibodies, bispecific T cell engagers, antibody drug conjugates and chimeric antigen receptor (CAR) T cells, have shown significant activity but come with unique toxicities and have implications for how treatment is sequenced. Advances in T-ALL have lagged behind those seen in B-ALL. However, agents such as nelarabine, bortezomib and CAR T cell therapy targeting T cell antigens have been examined with promising results seen. As our understanding of disease biology in ALL grows, as does our ability to target pathways such as apoptosis, through BH3 mimetics, chemokines and epigenetic regulators. This review aims to highlight a range of available and emerging targeted therapeutics in ALL, to explore their mechanisms of action and to discuss the current evidence for their use.
Collapse
Affiliation(s)
- Ross Salvaris
- Department of Clinical Haematology, Monash Health, Clayton 3168, Australia;
- School of Clinical Sciences at Monash Health, Monash University, Clayton 3168, Australia
| | - Pasquale Luke Fedele
- Department of Clinical Haematology, Monash Health, Clayton 3168, Australia;
- School of Clinical Sciences at Monash Health, Monash University, Clayton 3168, Australia
| |
Collapse
|
9
|
Shah NN, Sokol L. Targeting CD22 for the Treatment of B-Cell Malignancies. Immunotargets Ther 2021; 10:225-236. [PMID: 34262884 PMCID: PMC8275043 DOI: 10.2147/itt.s288546] [Citation(s) in RCA: 34] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2021] [Accepted: 06/24/2021] [Indexed: 01/17/2023] Open
Abstract
Immunotherapeutic agents play an increasingly important role in the treatment of B-cell malignancies. CD19 and CD20 are common targets for lymphoid malignancies, though patients who relapse have few therapeutic options remaining. CD22 is a cell surface sialoglycoprotein uniquely present on B-cells and regulates B-cell function and proliferation. Thus, it is an appealing therapeutic target for autoimmune disorders and B-cell malignancies. A variety of therapies targeting CD22 have been developed, including monoclonal antibodies, antibody-drug conjugates, radioimmunoconjugates, chimeric antigen receptor T cells, and bispecific antibodies. Here, we review the biology of CD22 and key therapies targeting CD22 in lymphoid malignancies.
Collapse
Affiliation(s)
- Nikesh N Shah
- Department of Internal Medicine, Morsani College of Medicine, University of South Florida, Tampa, FL, USA
| | - Lubomir Sokol
- Department of Malignant Hematology, H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL, USA
| |
Collapse
|
10
|
Pastorczak A, Domka K, Fidyt K, Poprzeczko M, Firczuk M. Mechanisms of Immune Evasion in Acute Lymphoblastic Leukemia. Cancers (Basel) 2021; 13:1536. [PMID: 33810515 PMCID: PMC8037152 DOI: 10.3390/cancers13071536] [Citation(s) in RCA: 34] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2021] [Revised: 03/17/2021] [Accepted: 03/22/2021] [Indexed: 02/07/2023] Open
Abstract
Acute lymphoblastic leukemia (ALL) results from a clonal expansion of abnormal lymphoid progenitors of B cell (BCP-ALL) or T cell (T-ALL) origin that invade bone marrow, peripheral blood, and extramedullary sites. Leukemic cells, apart from their oncogene-driven ability to proliferate and avoid differentiation, also change the phenotype and function of innate and adaptive immune cells, leading to escape from the immune surveillance. In this review, we provide an overview of the genetic heterogeneity and treatment of BCP- and T-ALL. We outline the interactions of leukemic cells in the bone marrow microenvironment, mainly with mesenchymal stem cells and immune cells. We describe the mechanisms by which ALL cells escape from immune recognition and elimination by the immune system. We focus on the alterations in ALL cells, such as overexpression of ligands for various inhibitory receptors, including anti-phagocytic receptors on macrophages, NK cell inhibitory receptors, as well as T cell immune checkpoints. In addition, we describe how developing leukemia shapes the bone marrow microenvironment and alters the function of immune cells. Finally, we emphasize that an immunosuppressive microenvironment can reduce the efficacy of chemo- and immunotherapy and provide examples of preclinical studies showing strategies for improving ALL treatment by targeting these immunosuppressive interactions.
Collapse
Affiliation(s)
- Agata Pastorczak
- Department of Pediatrics, Oncology and Hematology, Medical University of Lodz, 91-738 Lodz, Poland;
| | - Krzysztof Domka
- Department of Immunology, Medical University of Warsaw, 02-097 Warsaw, Poland; (K.D.); (K.F.); (M.P.)
- Doctoral School, Medical University of Warsaw, 02-091 Warsaw, Poland
| | - Klaudyna Fidyt
- Department of Immunology, Medical University of Warsaw, 02-097 Warsaw, Poland; (K.D.); (K.F.); (M.P.)
- Postgraduate School of Molecular Medicine, Medical University of Warsaw, 02-091 Warsaw, Poland
| | - Martyna Poprzeczko
- Department of Immunology, Medical University of Warsaw, 02-097 Warsaw, Poland; (K.D.); (K.F.); (M.P.)
| | - Malgorzata Firczuk
- Department of Immunology, Medical University of Warsaw, 02-097 Warsaw, Poland; (K.D.); (K.F.); (M.P.)
| |
Collapse
|
11
|
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
|
12
|
Jammal N, Chew S, Jabbour E, Kantarjian H. Antibody based therapy in relapsed acute lymphoblastic leukemia. Best Pract Res Clin Haematol 2020; 33:101225. [PMID: 33279181 DOI: 10.1016/j.beha.2020.101225] [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: 11/06/2020] [Accepted: 11/09/2020] [Indexed: 10/22/2022]
Abstract
Outcomes for relapsed and refractory acute lymphoblastic leukemia (ALL) remain poor. With the advent of targeted monoclonal antibodies and antibody constructs, these outcomes have been significantly improved both in the frontline and salvage setting. These targets include a bispecific antibody that targets both CD3 and CD19, known as blinatumomab, as well as a conjugated antibody that targets CD22, known as inotuzumab ozogamicin. These agents have been thoroughly studied and successively approved for use as monotherapy, however, more recently they have been incorporated in combination or sequentially with cytotoxic chemotherapy. In this chapter, we will discuss the role that these monoclonal antibodies play as monotherapy and in combination in the treatment of ALL in the salvage setting, and how they continue to transform the treatment management of relapsed and refractory ALL.
Collapse
Affiliation(s)
- Nadya Jammal
- Department of Leukemia, University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Serena Chew
- Department of Leukemia, University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Elias Jabbour
- Department of Leukemia, University of Texas MD Anderson Cancer Center, Houston, TX, USA.
| | - Hagop Kantarjian
- Department of Leukemia, University of Texas MD Anderson Cancer Center, Houston, TX, USA
| |
Collapse
|
13
|
Rank CU, Schmiegelow K. Optimal approach to the treatment of young adults with acute lymphoblastic leukemia in 2020. Semin Hematol 2020; 57:102-114. [PMID: 33256899 DOI: 10.1053/j.seminhematol.2020.08.001] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2020] [Revised: 08/12/2020] [Accepted: 08/14/2020] [Indexed: 01/19/2023]
Abstract
Akin to the introduction of tyrosine kinase inhibitors to Philadelphia chromosome-positive acute lymphoblastic leukemia (ALL), pediatric-based asparaginase-heavy approaches have revolutionized the treatment of young adults with the Philadelphia chromosome-negative subset the past decades. Once again, we are approaching a new era. An era of precision medicine with immunotherapy and other molecularly targeted treatments that offers unique opportunities to customize treatment intensity with or without hematopoietic stem cell transplantation, reduce the burden of toxicities, and combat persistent residual disease. Recently approved agents for refractory/relapsed B-cell precursor ALL include the chimeric antigen receptor-modified T-cells, the anti-CD22 monoclonal antibody-drug conjugate, inotuzumab ozogamicin, and the bispecific anti-CD19 T-cell engager, blinatumomab. These agents are expected to move widely into the frontline setting along with the proteasome inhibitors, bortezomib and carfilzomib, as well as tyrosine kinase inhibitors for Philadelphia-like rearrangements that are especially frequent among young adults. To this add the BH3 mimetics, venetoclax and navitoclax, which are being widely explored in refractory/relapsed as well as frontline settings for B- and T-cell ALL. The promising anti-CD38 monoclonal antibody, daratumumab, is entering the scene of refractory/relapsed T-ALL, whereas the old purine analogue, nelarabine, is being evaluated in a new upfront setting. This review focuses on 2 main questions: How do we optimize frontline as well as salvage ALL treatment of young adults in the 2020s? Not least, how do we address the current burden of serious toxicities unique to young adults?
Collapse
Affiliation(s)
- Cecilie Utke Rank
- Pediatric Oncology Research Laboratory, Rigshospitalet, University of Copenhagen, Copenhagen, Denmark; Department of Hematology, Rigshospitalet, University of Copenhagen, Copenhagen, Denmark
| | - Kjeld Schmiegelow
- Pediatric Oncology Research Laboratory, Rigshospitalet, University of Copenhagen, Copenhagen, Denmark; Department of Pediatrics and Adolescent Medicine, Rigshospitalet, University of Copenhagen, Copenhagen, Denmark; Institute of Clinical Medicine, Faculty of Medicine, University of Copenhagen, Copenhagen, Denmark.
| |
Collapse
|
14
|
Ribera JM, Genescà E, Ribera J. Bispecific T-cell engaging antibodies in B-cell precursor acute lymphoblastic leukemias: focus on blinatumomab. Ther Adv Hematol 2020; 11:2040620720919632. [PMID: 32523659 PMCID: PMC7236391 DOI: 10.1177/2040620720919632] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2019] [Accepted: 03/05/2020] [Indexed: 11/28/2022] Open
Abstract
Bispecific T-cell engaging antibodies are constructs engineered to bind to two different antigens, one to a tumor-specific target and the other to CD3-positive T cells or natural killer (NK) cells. Blinatumomab engages CD19 and CD3, performing effective serial lysis. The clinical development program in acute lymphoblastic leukemia (ALL) includes clinical trials in relapsed or refractory (R/R) patients and in B-cell precursor (BCP) ALL patients with measurable residual disease. Several trials are currently being conducted in de novo BCP-ALL, either in induction, consolidation, or before or after hematopoietic stem cell transplant. Combination with other targeted therapies or with other immunotherapeutic approaches are also underway. Several strategies are aimed to optimize the use of blinatumomab either by overcoming the mechanisms of resistance (e.g. inhibition of PD-1/PD-L1) or by improvements in the route of application, among others.
Collapse
Affiliation(s)
- Jose-Maria Ribera
- Clinical Hematology Department, ICO-Hospital Germans Trias I Pujol, Josep Carreras Research Institute, Universitat Autònoma de Barcelona, C/ Canyet, s/n, Badalona, 08916, Spain
| | - Eulalia Genescà
- Clinical Hematology Department, ICO-Hospital Germans Trias i Pujol, Josep Carreras Research Institute, Universitat Autònoma de Barcelona, Badalona, Spain
| | - Jordi Ribera
- Clinical Hematology Department, ICO-Hospital Germans Trias i Pujol, Josep Carreras Research Institute, Universitat Autònoma de Barcelona, Badalona, Spain
| |
Collapse
|
15
|
Foster JC, Freidlin B, Kunos CA, Korn EL. Single-Arm Phase II Trials of Combination Therapies: A Review of the CTEP Experience 2008-2017. J Natl Cancer Inst 2020; 112:128-135. [PMID: 31545373 PMCID: PMC7019095 DOI: 10.1093/jnci/djz193] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2019] [Revised: 08/28/2019] [Accepted: 09/13/2019] [Indexed: 01/08/2023] Open
Abstract
Designing and interpreting single-arm phase II trials of combinations of agents is challenging because it can be difficult, based on historical data, to identify levels of activity for which the combination would be worth pursuing. We identified Cancer Therapy Evaluation Program single-arm combination trials that were activated in 2008-2017 and tabulated their design characteristics and results. Positive trials were evaluated as to whether they provided credible evidence that the combination was better than its constituents. A total of 125 trials were identified, and 120 trials had results available. Twelve had designs where eligible patients were required to be resistant or refractory to all but one element of the combination. Only 17.8% of the 45 positive trials were deemed to provide credible evidence that the combination was better than its constituents. Of the 10 positive trials with observed rates 10 percentage points higher than their upper (alternative hypothesis) targets, only five were deemed to provide such credible evidence. Many trials were definitively negative, with observed clinical activity at or below their lower (null hypothesis) targets. Ideally, use of single-arm combination trials should be restricted to settings where each agent is known to have minimal monotherapy activity (and a randomized trial is infeasible). In these settings, an observed signal is attributable to synergy and thus could be used to decide whether the combination is worth pursuing. In other settings, credible evidence can still be obtained if the observed activity is much higher than expected, but experience suggests that this is a rare occurrence.
Collapse
Affiliation(s)
- Jared C Foster
- Biometric Research Program, Division of Cancer Treatment and Diagnosis, National Cancer Institute, Bethesda, MD
| | - Boris Freidlin
- Biometric Research Program, Division of Cancer Treatment and Diagnosis, National Cancer Institute, Bethesda, MD
| | - Charles A Kunos
- Cancer Therapy Evaluation Program, Division of Cancer Treatment and Diagnosis, National Cancer Institute, Bethesda, MD
| | - Edward L Korn
- Biometric Research Program, Division of Cancer Treatment and Diagnosis, National Cancer Institute, Bethesda, MD
| |
Collapse
|
16
|
Deak D, Pop C, Zimta AA, Jurj A, Ghiaur A, Pasca S, Teodorescu P, Dascalescu A, Antohe I, Ionescu B, Constantinescu C, Onaciu A, Munteanu R, Berindan-Neagoe I, Petrushev B, Turcas C, Iluta S, Selicean C, Zdrenghea M, Tanase A, Danaila C, Colita A, Colita A, Dima D, Coriu D, Einsele H, Tomuleasa C. Let's Talk About BiTEs and Other Drugs in the Real-Life Setting for B-Cell Acute Lymphoblastic Leukemia. Front Immunol 2020; 10:2856. [PMID: 31921126 PMCID: PMC6934055 DOI: 10.3389/fimmu.2019.02856] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2019] [Accepted: 11/20/2019] [Indexed: 01/07/2023] Open
Abstract
Background: Therapy for acute lymphoblastic leukemia (ALL) are currently initially efficient, but even if a high percentage of patients have an initial complete remission (CR), most of them relapse. Recent data shows that immunotherapy with either bispecific T-cell engagers (BiTEs) of chimeric antigen receptor (CAR) T cells can eliminate residual chemotherapy-resistant B-ALL cells. Objective: The objective of the manuscript is to present improvements in the clinical outcome for chemotherapy-resistant ALL in the real-life setting, by describing Romania's experience with bispecific antibodies for B-cell ALL. Methods: We present the role of novel therapies for relapsed B-cell ALL, including the drugs under investigation in phase I-III clinical trials, as a potential bridge to transplant. Blinatumomab is presented in a critical review, presenting both the advantages of this drug, as well as its limitations. Results: Bispecific antibodies are discussed, describing the clinical trials that resulted in its approval by the FDA and EMA. The real-life setting for relapsed B-cell ALL is described and we present the patients treated with blinatumomab in Romania. Conclusion: In the current manuscript, we present blinatumomab as a therapeutic alternative in the bridge-to-transplant setting for refractory or relapsed ALL, to gain a better understanding of the available therapies and evidence-based data for these patients in 2019.
Collapse
Affiliation(s)
- Dalma Deak
- Department of Hematology, Iuliu Hatieganu University of Medicine and Pharmacy, Cluj-Napoca, Romania.,Department of Hematology, Ion Chiricuta Clinical Cancer Center, Cluj-Napoca, Romania
| | - Cristina Pop
- Department of Pharmacology, Faculty of Pharmacy, Iuliu Hatieganu University of Medicine and Pharmacy, Cluj-Napoca, Romania
| | - Alina-Andreea Zimta
- Research Center for Advanced Medicine, Iuliu Hatieganu University of Medicine and Pharmacy, Cluj-Napoca, Romania
| | - Ancuta Jurj
- Research Center for Functional Genomics and Translational Medicine, Iuliu Hatieganu University of Medicine and Pharmacy, Cluj-Napoca, Romania
| | - Alexandra Ghiaur
- Department of Hematology, Fundeni Clinical Institute, Bucharest, Romania
| | - Sergiu Pasca
- Department of Hematology, Iuliu Hatieganu University of Medicine and Pharmacy, Cluj-Napoca, Romania
| | - Patric Teodorescu
- Department of Hematology, Iuliu Hatieganu University of Medicine and Pharmacy, Cluj-Napoca, Romania
| | - Angela Dascalescu
- Department of Hematology, Grigore T. Popa University of Medicine and Pharmacy, Iasi, Romania.,Department of Hematology, Regional Institute of Oncology, Iasi, Romania
| | - Ion Antohe
- Department of Hematology, Grigore T. Popa University of Medicine and Pharmacy, Iasi, Romania.,Department of Hematology, Regional Institute of Oncology, Iasi, Romania
| | - Bogdan Ionescu
- Department of Hematology, Fundeni Clinical Institute, Bucharest, Romania
| | - Catalin Constantinescu
- Department of Hematology, Iuliu Hatieganu University of Medicine and Pharmacy, Cluj-Napoca, Romania
| | - Anca Onaciu
- Department of Hematology, Iuliu Hatieganu University of Medicine and Pharmacy, Cluj-Napoca, Romania
| | - Raluca Munteanu
- Department of Hematology, Iuliu Hatieganu University of Medicine and Pharmacy, Cluj-Napoca, Romania
| | - Ioana Berindan-Neagoe
- Department of Hematology, Iuliu Hatieganu University of Medicine and Pharmacy, Cluj-Napoca, Romania
| | - Bobe Petrushev
- Department of Hematology, Iuliu Hatieganu University of Medicine and Pharmacy, Cluj-Napoca, Romania
| | - Cristina Turcas
- Department of Hematology, Iuliu Hatieganu University of Medicine and Pharmacy, Cluj-Napoca, Romania.,Department of Hematology, Ion Chiricuta Clinical Cancer Center, Cluj-Napoca, Romania
| | - Sabina Iluta
- Department of Hematology, Iuliu Hatieganu University of Medicine and Pharmacy, Cluj-Napoca, Romania
| | - Cristina Selicean
- Department of Hematology, Iuliu Hatieganu University of Medicine and Pharmacy, Cluj-Napoca, Romania
| | - Mihnea Zdrenghea
- Department of Hematology, Iuliu Hatieganu University of Medicine and Pharmacy, Cluj-Napoca, Romania.,Department of Hematology, Ion Chiricuta Clinical Cancer Center, Cluj-Napoca, Romania
| | - Alina Tanase
- Department of Stem Cell Transplantation, Fundeni Clinical Institute, Bucharest, Romania
| | - Catalin Danaila
- Department of Hematology, Grigore T. Popa University of Medicine and Pharmacy, Iasi, Romania.,Department of Hematology, Regional Institute of Oncology, Iasi, Romania
| | - Anca Colita
- Department of Stem Cell Transplantation, Fundeni Clinical Institute, Bucharest, Romania.,Department of Pediatrics, Carol Davila University of Medicine and Pharmacy, Bucharest, Romania
| | - Andrei Colita
- Department of Hematology, Coltea Hospital, Bucharest, Romania.,Department of Hematology, Carol Davila University of Medicine and Pharmacy, Bucharest, Romania
| | - Delia Dima
- Department of Hematology, Iuliu Hatieganu University of Medicine and Pharmacy, Cluj-Napoca, Romania
| | - Daniel Coriu
- Department of Hematology, Ion Chiricuta Clinical Cancer Center, Cluj-Napoca, Romania.,Department of Hematology, Fundeni Clinical Institute, Bucharest, Romania.,Department of Hematology, Carol Davila University of Medicine and Pharmacy, Bucharest, Romania
| | - Hermann Einsele
- Department of Internal Medicine II, University Hospital Wurzburg, Würzburg, Germany
| | - Ciprian Tomuleasa
- Department of Hematology/Research Center for Functional Genomics and Translational Medicine, Iuliu Hatieganu University of Medicine and Pharmacy, Cluj-Napoca, Romania
| |
Collapse
|
17
|
Shang Y, Zhou F. Current Advances in Immunotherapy for Acute Leukemia: An Overview of Antibody, Chimeric Antigen Receptor, Immune Checkpoint, and Natural Killer. Front Oncol 2019; 9:917. [PMID: 31616632 PMCID: PMC6763689 DOI: 10.3389/fonc.2019.00917] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2019] [Accepted: 09/03/2019] [Indexed: 12/13/2022] Open
Abstract
Recently, due to the application of hematopoietic stem cell transplantation and small molecule inhibitor, the survival of acute leukemia is prolonged. However, the 5 year survival rate remains low due to a high incidence of relapse. Immunotherapy is expected to improve the prognosis of patients with relapsed or refractory hematological malignancies because it does not rely on the cytotoxic mechanisms of conventional therapy. In this paper, the advances of immunotherapy in acute leukemia are reviewed from the aspects of Antibody including Unconjugated antibodies, Antibody-drug conjugate and Bispecific antibody, Chimeric Antigen Receptor (CARs), Immune checkpoint, Natural killer cells. The immunological features, mechanisms and limitation in clinic will be described.
Collapse
Affiliation(s)
- Yufeng Shang
- Department of Hematology, Zhongnan Hospital of Wuhan University, Wuhan, China
| | - Fuling Zhou
- Department of Hematology, Zhongnan Hospital of Wuhan University, Wuhan, China
| |
Collapse
|
18
|
Maffini E, Saraceni F, Lanza F. Treatment of Adult Patients with Relapsed/Refractory B-Cell Philadelphia-Negative Acute Lymphoblastic Leukemia. Clin Hematol Int 2019; 1:85-93. [PMID: 34595415 PMCID: PMC8432388 DOI: 10.2991/chi.d.190503.002] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2019] [Accepted: 04/26/2019] [Indexed: 12/27/2022] Open
Abstract
The majority of adult patients affected by B-cell acute lymphoblastic leukemia (B-ALL) will relapse after an initial response, while approximately 20% will display primary resistant disease. Patients suffering from relapsed/refractory B-ALL have a very poor outcome. Allogeneic hematopoietic cell transplantation (HCT) still represents the only curative approach, but is not so frequently feasible, because of patient's fitness, donor availability, and the ability to achieve a remission prior to HCT. The estimated remission rates with conventional cytotoxic agents are around 30%, but they are short-lived. These disappointing results led to the introduction of new immunologic-based treatments-blinatumomab and inotuzumab. They produced a substantial improvement in terms of response rates, with the ability, in most cases, to induce a minimal residual disease (MRD)-negative status. Similarly, T cells engineered to express a CD19-specific chimeric antigen receptor (CAR-T) have yielded sensational results among patients with relapsed/refractory B-ALL, with unexpectedly high MRD-negative complete remissions rates. However, the first studies looking at long-term outcomes after CAR-T infusions told us that a significant fraction of such responses are not durable, and may benefit from a consolidation approach such as an allogeneic HCT.
Collapse
Affiliation(s)
- Enrico Maffini
- Hematology Unit, Romagna Transplant Network, Hospital of Ravenna, Viale Randi n. 5, 48121 Ravenna, Italy
| | - Francesco Saraceni
- Hematology Unit, Romagna Transplant Network, Hospital of Ravenna, Viale Randi n. 5, 48121 Ravenna, Italy
| | - Francesco Lanza
- Hematology Unit, Romagna Transplant Network, Hospital of Ravenna, Viale Randi n. 5, 48121 Ravenna, Italy
| |
Collapse
|
19
|
Guerra VA, Jabbour EJ, Ravandi F, Kantarjian H, Short NJ. Novel monoclonal antibody-based treatment strategies in adults with acute lymphoblastic leukemia. Ther Adv Hematol 2019; 10:2040620719849496. [PMID: 31205644 PMCID: PMC6535741 DOI: 10.1177/2040620719849496] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2019] [Accepted: 04/18/2019] [Indexed: 12/21/2022] Open
Abstract
Adult acute lymphoblastic leukemia (ALL) has a poor overall survival compared with pediatric ALL where cure rates are observed in more than 90% of patients. The recent development of novel monoclonal antibodies targeting CD20, CD19, and CD22 has changed the long-term outcome of this disease, both in the frontline setting (e.g. rituximab) and for patients with relapsed/refractory disease (e.g. inotuzumab ozogamicin and blinatumomab). The CD3-CD19 bispecific T-cell-engaging antibody blinatumomab is also the first drug approved in ALL for patients with persistent or recurrent measurable residual disease, providing a new treatment paradigm for these patients. Several new agents are also in development that use novel constructs or target alternative surface epitopes such as CD123, CD25, and CD38. Herein, we review the role of monoclonal antibodies in adult ALL and summarize the current and future approaches in ALL, including novel combination therapies and the possibility of early incorporation of these agents into treatment regimens.
Collapse
Affiliation(s)
- Veronica A Guerra
- Department of Leukemia, University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Elias J Jabbour
- Department of Leukemia, University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Farhad Ravandi
- Department of Leukemia, University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Hagop Kantarjian
- Department of Leukemia, University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Nicholas J Short
- Department of Leukemia, Unit 428, The University of Texas MD Anderson Cancer Center, 1515 Holcombe Boulevard, Houston, TX 77030, USA
| |
Collapse
|
20
|
Abstract
PURPOSE OF REVIEW Treatment options for patients with acute lymphoblastic leukemia (ALL) beyond standard chemotherapy have grown significantly in recent years. In this review, we highlight new targeted therapies in ALL, with an emphasis on immunotherapy. RECENT FINDINGS Major advances include antibody-based therapies, such as naked monoclonal antibodies, antibody-drug conjugates and bispecific T cell engaging (BiTE) antibodies, as well as adoptive cellular therapies such as chimeric antigen receptor (CAR) T cells. Apart from the above immunotherapeutic approaches, other targeted therapies are being employed in Philadelphia chromosome-positive (Ph+) ALL, Philadelphia-like (Ph-like) ALL, and T cell ALL. These new treatment strategies are changing the treatment landscape of ALL and challenging the current standard of care. Clinical trials will hopefully help determine how to best incorporate these novel therapies into existing treatment algorithms.
Collapse
Affiliation(s)
- Kathleen W Phelan
- Cardinal Bernardin Cancer Center, Loyola University Medical Center, 2160 S. First Avenue, Maywood, IL, 60153, USA
| | - Anjali S Advani
- Taussig Cancer Center, Cleveland Clinic, 10201 Carnegie Avenue, Desk CA60, Cleveland, OH, 44195, USA.
| |
Collapse
|
21
|
Mohseni M, Uludag H, Brandwein JM. Advances in biology of acute lymphoblastic leukemia (ALL) and therapeutic implications. AMERICAN JOURNAL OF BLOOD RESEARCH 2018; 8:29-56. [PMID: 30697448 PMCID: PMC6334189] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Received: 11/14/2018] [Accepted: 12/06/2018] [Indexed: 06/09/2023]
Abstract
Acute lymphoblastic leukemia (ALL) is the most common pediatric cancer and also occurs in adults. Although the outcomes of multi-agent chemotherapy regimens have greatly improved, high toxicity and relapses in many patients necessitate the development of novel therapeutic approaches. Advances in molecular profiling and cytogenetics have identified a broad range of genetic abnormalities, including gene mutations, chromosome translocations and aneuploidy, which has provided a more comprehensive understanding of the biology and pathogenesis of ALL. This understanding has also led to new targeted therapeutic approaches, including the use of selective small molecule inhibitors, nucleic acid-based therapies and immune-based therapies mediated by specific monoclonal antibodies and cellular immunotherapy, which are poised to revolutionize the treatment of various ALL subtypes. The main focus of this review is to highlight the latest advances in ALL biology, including the identification of prognostic factors and putative therapeutic targets. We also review the current status of, and ongoing progress in, the development of targeted therapies for ALL.
Collapse
Affiliation(s)
- Mahsa Mohseni
- Department of Medicine, University of Alberta Edmonton, Alberta, Canada
| | - Hasan Uludag
- Department of Chemical and Materials Engineering, University of Alberta Edmonton, Alberta, Canada
| | | |
Collapse
|
22
|
Dinner S, Liedtke M. Antibody-based therapies in patients with acute lymphoblastic leukemia. HEMATOLOGY. AMERICAN SOCIETY OF HEMATOLOGY. EDUCATION PROGRAM 2018; 2018:9-15. [PMID: 30504286 PMCID: PMC6246018 DOI: 10.1182/asheducation-2018.1.9] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/04/2023]
Abstract
The use of multiagent combination chemotherapy regimens results in cure rates of >90% for children and ∼40% for adults with acute lymphoblastic leukemia (ALL) but is associated with extensive toxicity and disappointingly low efficacy in relapsed patients. ALL blast cells express several surface antigens, including CD20, CD22, and CD19, which represent valuable targets for immunotherapy. Monoclonal antibodies, antibody-drug conjugates, and bispecific T-cell-engaging antibodies targeting these antigens offer novel mechanisms of action. Within the last several years, the anti-CD20 antibody rituximab has been added to chemotherapy for newly diagnosed patients <60 years with CD20+ pre-B ALL and significantly improved the 2-year event-free survival from 52% to 65%. In adults with relapsed or refractory CD22+ ALL, the antibody-drug conjugate inotuzumab ozogamicin resulted in a complete response rate of 81% and median overall survival of 7.7 months with reduced toxicity compared with standard chemotherapy. Similarly, the bispecific T-cell-engaging antibody blinatumomab yielded a complete response rate of 44% and a median overall survival of 7.7 months in an extensively treated ALL population. Moreover, ∼80% of ALL patients in complete remission with evidence of minimal residual disease (MRD) achieved a complete MRD response following treatment with blinatumomab. These results highlight the tremendous promise of antibody-based treatment approaches for ALL. Ongoing and future research is critical to further define the role of the various immunotherapies in the frontline treatment of ALL. Additional challenges include the optimal sequencing of the available antibodies in the relapsed setting as well as their integration with stem cell transplant and chimeric antigen receptor T-cell therapy.
Collapse
Affiliation(s)
- Shira Dinner
- Division of Hematology/Oncology, Department of Medicine, Northwestern University Feinberg School of Medicine, Chicago, IL; and
| | - Michaela Liedtke
- Division of Hematology, Department of Medicine, Stanford University School of Medicine, Stanford, CA
| |
Collapse
|
23
|
Bassan R, Bourquin JP, DeAngelo DJ, Chiaretti S. New Approaches to the Management of Adult Acute Lymphoblastic Leukemia. J Clin Oncol 2018; 36:JCO2017773648. [PMID: 30240326 DOI: 10.1200/jco.2017.77.3648] [Citation(s) in RCA: 63] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/20/2023] Open
Abstract
Traditional treatment regimens for adult acute lymphoblastic leukemia, including allogeneic hematopoietic cell transplantation, result in an overall survival of approximately 40%, a figure hardly comparable with the extraordinary 80% to 90% cure rate currently reported in children. When translated to the adult setting, modern pediatric-type regimens improve the survival to approximately 60% in young adults. The addition of tyrosine kinase inhibitors for patients with Philadelphia chromosome-positive disease and the measurement of minimal residual disease to guide risk stratification and postremission approaches has led to additional improvements in outcomes. Relapsed disease and treatment toxicity-sparing no patient but representing a major concern especially in the elderly-are the most critical current issues awaiting further therapeutic advancement. Recently, there has been considerable progress in understanding the disease biology, specifically the Philadelphia-like signature, as well as other high-risk subgroups. In addition, there are several new agents that will undoubtedly contribute to additional improvement in the current outcomes. The most promising agents are monoclonal antibodies, immunomodulators, and chimeric antigen receptor T cells, and, to a lesser extent, several new drugs targeting key molecular pathways involved in leukemic cell growth and proliferation. This review examines the evidence supporting the increasing role of the new therapeutic tools and treatment options in different disease subgroups, including frontline and relapsed or refractory disease. It is now possible to define the best individual approach on the basis of the emerging concepts of precision medicine.
Collapse
Affiliation(s)
- Renato Bassan
- Renato Bassan, Ospedale dell'Angelo, Mestre-Venezia; Sabina Chiaretti, "Sapienza" University, Rome, Italy; Jean-Pierre Bourquin, University Children's Hospital, Zurich, Switzerland; and Daniel J. DeAngelo, Dana-Farber Cancer Institute, Boston, MA
| | - Jean-Pierre Bourquin
- Renato Bassan, Ospedale dell'Angelo, Mestre-Venezia; Sabina Chiaretti, "Sapienza" University, Rome, Italy; Jean-Pierre Bourquin, University Children's Hospital, Zurich, Switzerland; and Daniel J. DeAngelo, Dana-Farber Cancer Institute, Boston, MA
| | - Daniel J DeAngelo
- Renato Bassan, Ospedale dell'Angelo, Mestre-Venezia; Sabina Chiaretti, "Sapienza" University, Rome, Italy; Jean-Pierre Bourquin, University Children's Hospital, Zurich, Switzerland; and Daniel J. DeAngelo, Dana-Farber Cancer Institute, Boston, MA
| | - Sabina Chiaretti
- Renato Bassan, Ospedale dell'Angelo, Mestre-Venezia; Sabina Chiaretti, "Sapienza" University, Rome, Italy; Jean-Pierre Bourquin, University Children's Hospital, Zurich, Switzerland; and Daniel J. DeAngelo, Dana-Farber Cancer Institute, Boston, MA
| |
Collapse
|
24
|
Statler A, Othus M, Erba HP, Chauncey TR, Radich JP, Coutre S, Advani A, Nand S, Ravandi F, Mukherjee S, Sekeres MA. Comparable outcomes of patients eligible vs ineligible for SWOG leukemia studies. Blood 2018; 131:2782-2788. [PMID: 29618479 PMCID: PMC6014358 DOI: 10.1182/blood-2018-01-826693] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2018] [Accepted: 03/30/2018] [Indexed: 02/03/2023] Open
Abstract
Patients may be deemed ineligible for a clinical trial for reasons that do not directly impact efficacy or safety. We identified reasons for ineligibility and compared outcomes of ineligible with eligible patients treated on Southwest Oncology Group (SWOG) Leukemia Committee protocols. Patients enrolled in SWOG phase 2, 2/3, or 3 protocols open since 2005 were analyzed for eligibility status, reasons for ineligibility, baseline characteristics, Eastern Cooperative Oncology Group (ECOG) performance status (PS), serious adverse events (SAEs), complete remission (CR) status, and overall survival. A total of 2361 patients were enrolled in the 13 included studies. Of these, 247 (10%) were deemed ineligible; 78 were excluded from analyses, and 169 were included. Of the 169 included in analyses, 60% (101/169) were excluded due to missing baseline documentation. Baseline characteristics comparing ineligible to eligible patients were similar, with the exception of ECOG PS for S0325 (P = .02) and S0530 (P = .002). In multivariable analyses, neither the proportion of patients with ECOG PS ≥ 2 (P = .12) nor the rate of grade 5 SAEs (P = .62) differed between groups. There was no difference in survival between eligible and ineligible patients (P = .25), and CR rates were similar, with the exception of S0325 (P < .001) and S0703 (P = .004). The findings of this study suggest that nonessential eligibility criteria can be less restrictive, thus expanding patient enrollment and avoiding protocol deviations. The clinical trials included in this study were registered at www.clincialtrials.gov as #NCT00085709, #NCT00066794, #NCT00070499, #NCT00109837, #NCT00093418, #NCT00492856, #NCT00337168, #NCT00352365, #NCT00658814, #NCT00792948, #NCT00945815, #NCT00840177, and #NCT01522976.
Collapse
Affiliation(s)
- Abby Statler
- Leukemia Program, Taussig Cancer Institute, Cleveland Clinic, Cleveland, OH
| | - Megan Othus
- Fred Hutchinson Cancer Research Center, Seattle, WA
| | - Harry P Erba
- SWOG Leukemia Committee, Portland, OR
- Division of Hematology and Oncology, University of Alabama, Birmingham, AL
| | - Thomas R Chauncey
- Clinical Research Division, Fred Hutchinson Cancer Research Center, Seattle, WA
| | - Jerald P Radich
- Clinical Research Division, Fred Hutchinson Cancer Research Center, Seattle, WA
| | - Steven Coutre
- Stanford Cancer Center, Stanford University School of Medicine, Stanford, CA
| | - Anjali Advani
- Leukemia Program, Taussig Cancer Institute, Cleveland Clinic, Cleveland, OH
| | - Sucha Nand
- Division of Hematology/Oncology, Department of Medicine, Loyola University Medical Center, Maywood, IL; and
| | - Farhad Ravandi
- Department of Leukemia, The University of Texas MD Anderson Cancer Center, Houston, TX
| | - Sudipto Mukherjee
- Leukemia Program, Taussig Cancer Institute, Cleveland Clinic, Cleveland, OH
| | - Mikkael A Sekeres
- Leukemia Program, Taussig Cancer Institute, Cleveland Clinic, Cleveland, OH
| |
Collapse
|
25
|
Drgona L, Gudiol C, Lanini S, Salzberger B, Ippolito G, Mikulska M. 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 or myeloid cells surface antigens [II]: CD22, CD30, CD33, CD38, CD40, SLAMF-7 and CCR4). Clin Microbiol Infect 2018; 24 Suppl 2:S83-S94. [DOI: 10.1016/j.cmi.2018.03.022] [Citation(s) in RCA: 71] [Impact Index Per Article: 10.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2017] [Revised: 02/05/2018] [Accepted: 02/11/2018] [Indexed: 01/12/2023]
|
26
|
Abstract
Chemotherapy cures only a minority of adult patients with acute lymphoblastic leukemia (ALL). In addition, relapsed ALL has a poor outcome with 5-year survival as low as 7 %. Hence, there is a need to develop effective therapies to treat relapsed disease and to combine these agents with chemotherapy to improve outcomes in newly diagnosed patients. ALL cells express several antigens amenable to target therapies including CD19, CD20, CD22, and CD52. Over the last decade, there has been a surge in the development of immune therapies which target these receptors and that have induced robust responses. In this manuscript, we review these novel immune agents in the treatment of B-ALL. As these new therapies mature, the challenge going forward will be to find safe and effective combinations of these agents with chemotherapy and to determine their place in the current treatment schema.
Collapse
|
27
|
Wei G, Wang J, Huang H, Zhao Y. Novel immunotherapies for adult patients with B-lineage acute lymphoblastic leukemia. J Hematol Oncol 2017; 10:150. [PMID: 28821272 PMCID: PMC5563021 DOI: 10.1186/s13045-017-0516-x] [Citation(s) in RCA: 68] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2017] [Accepted: 08/07/2017] [Indexed: 12/16/2022] Open
Abstract
The past decade witnessed the rapid development of adult B-lineage acute lymphoblastic leukemia (ALL) treatment. Beyond the development of chemotherapy regimens, immunotherapy is starting a new era with unprecedented complete remission (CR) rate. Targeting B-lineage-specific surface markers such as CD19, CD20, CD22, or CD52, immunotherapy has been demonstrating promising clinical results. Among the immunotherapeutic methods, naked monoclonal antibodies (mAbs), antibody-drug conjugate (ADC), bispecific T cell engager (BiTE), and chimeric antigen receptor (CAR) T cells are the main types. In this review, we will examine the emerging preclinical and clinical development on (1) anti-CD20 naked mAbs rituximab, ofatumumab, and obinutuzumab; (2) anti-CD19 ADCs SAR3419 and SGN-CD19A and anti-CD19 BiTE blinatumomab; (3) anti-CD22 naked mAb epratuzumab and anti-CD22 ADC inotuzumab ozogamicin; (4) anti-CD52 naked mAb alemtuzumab; and (5) anti-CD19 CAR T cells. We will discuss their efficacy, adverse effects, as well as future development.
Collapse
Affiliation(s)
- Guoqing Wei
- Bone Marrow Transplantation Center, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, 310000, China
| | - Jiasheng Wang
- Bone Marrow Transplantation Center, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, 310000, China
| | - He Huang
- Bone Marrow Transplantation Center, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, 310000, China
| | - Yanmin Zhao
- Bone Marrow Transplantation Center, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, 310000, China.
| |
Collapse
|
28
|
Ronson A, Tvito A, Rowe JM. Treatment of Relapsed/Refractory Acute Lymphoblastic Leukemia in Adults. Curr Oncol Rep 2017; 18:39. [PMID: 27207612 DOI: 10.1007/s11912-016-0519-8] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023]
Abstract
Patients with relapsed and refractory acute lymphoblastic leukemia (ALL) have a dismal prognosis with less than 10 % of patients surviving 5 years. Most such patients cannot be rescued with currently available therapies, whatever the initial treatment they receive. Therefore, there is an urgent need for novel treatment options. Fortunately, over the past several years, an improved understanding of the biology of the disease has allowed the identification of rational molecular targets for therapeutic endeavors and the emergence of novel therapies has sparked great interest. This review will discuss the current treatment landscape for adult patients with relapsed and/or refractory ALL.
Collapse
Affiliation(s)
- Aharon Ronson
- Department of Hematology, Shaare Zedek Medical Center, 12 Shmuel Bayit Street, Jerusalem, 91031, Israel
| | - Ariella Tvito
- Department of Hematology, Shaare Zedek Medical Center, 12 Shmuel Bayit Street, Jerusalem, 91031, Israel
| | - Jacob M Rowe
- Department of Hematology, Shaare Zedek Medical Center, 12 Shmuel Bayit Street, Jerusalem, 91031, Israel. .,Technion, Israel Institute of Technology, Haifa, Israel.
| |
Collapse
|
29
|
Gökbuget N. How should we treat a patient with relapsed Ph-negative B-ALL and what novel approaches are being investigated? Best Pract Res Clin Haematol 2017; 30:261-274. [PMID: 29050699 DOI: 10.1016/j.beha.2017.07.010] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2017] [Revised: 07/29/2017] [Accepted: 07/31/2017] [Indexed: 12/25/2022]
Abstract
Despite significant improvements in outcome of newly diagnosed B-precursor ALL, the results in relapsed or refractory adult ALL are overall poor. Large retrospective studies revealed significant differences in terms of outcome, with particularly poor response rates in early or refractory relapses, whereas late relapses usually respond very well to repeated standard induction. Particularly new immunotherapy compounds like the CD19 bispecific antibody Blinatumomab and the conjugated CD22 antibody Inotuzumab yielded promising response rates compared to standard therapies in randomised trials. Long-term survival is however still poor. The optimal use of these compounds remains to be defined. Chimeric antigen receptor T-cells are another promising treatment approach and multicenter clinical trials in adult ALL are awaited. For selected patients molecular directed therapies may have a role in relapsed ALL; standard diagnostic algorithms need to be defined. One of the major challenges is to define the role of stem cell transplantation after relapse. Whereas this procedure appears to be the only chance for cure, the mortality and relapse rate are still high and optimisation is urgently needed. Future strategies include optimised use of new compounds as part of combination regimens and the earlier treatment of upcoming relapse in the situation of persistent or recurrent minimal residual disease.
Collapse
Affiliation(s)
- Nicola Gökbuget
- Universitätsklinikum Frankfurt/Main, Medizinische Klinik II, Hämatologie/Onkologie, Theodor-Stern-Kai 7, 60590, Frankfurt, Germany.
| |
Collapse
|
30
|
Saygin C, Papadantonakis N, Cassaday RD, Liedtke M, Fischer K, Dunn T, Patel BJ, Sobecks R, Kalaycio M, Sekeres MA, Mukherjee S, Gerds AT, Hamilton BK, Carraway HE, Advani AS. Prognostic impact of incomplete hematologic count recovery and minimal residual disease on outcome in adult acute lymphoblastic leukemia at the time of second complete response. Leuk Lymphoma 2017; 59:363-371. [DOI: 10.1080/10428194.2017.1344842] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/03/2023]
Affiliation(s)
- Caner Saygin
- Department of Hematology/Oncology, Taussig Cancer Institute, Cleveland Clinic, Cleveland, OH, USA
| | - Nikolaos Papadantonakis
- Department of Hematology/Oncology, Taussig Cancer Institute, Cleveland Clinic, Cleveland, OH, USA
| | - Ryan D. Cassaday
- Division of Hematology, Department of Medicine, University of Washington School of Medicine, and Clinical Research Division, Fred Hutchinson Cancer Research Center, Seattle, WA, USA
| | - Michaela Liedtke
- Department of Medicine, Stanford University School of Medicine, Stanford, CA, USA
| | - Katrina Fischer
- Department of Medicine, Stanford University School of Medicine, Stanford, CA, USA
| | - Tamara Dunn
- Department of Medicine, Stanford University School of Medicine, Stanford, CA, USA
| | - Bhumika J. Patel
- Department of Hematology/Oncology, Taussig Cancer Institute, Cleveland Clinic, Cleveland, OH, USA
| | - Ronald Sobecks
- Department of Hematology/Oncology, Taussig Cancer Institute, Cleveland Clinic, Cleveland, OH, USA
| | - Matt Kalaycio
- Department of Hematology/Oncology, Taussig Cancer Institute, Cleveland Clinic, Cleveland, OH, USA
| | - Mikkael A. Sekeres
- Department of Hematology/Oncology, Taussig Cancer Institute, Cleveland Clinic, Cleveland, OH, USA
| | - Sudipto Mukherjee
- Department of Hematology/Oncology, Taussig Cancer Institute, Cleveland Clinic, Cleveland, OH, USA
| | - Aaron T. Gerds
- Department of Hematology/Oncology, Taussig Cancer Institute, Cleveland Clinic, Cleveland, OH, USA
| | - Betty K. Hamilton
- Department of Hematology/Oncology, Taussig Cancer Institute, Cleveland Clinic, Cleveland, OH, USA
| | - Hetty E. Carraway
- Department of Hematology/Oncology, Taussig Cancer Institute, Cleveland Clinic, Cleveland, OH, USA
| | - Anjali S. Advani
- Department of Hematology/Oncology, Taussig Cancer Institute, Cleveland Clinic, Cleveland, OH, USA
| |
Collapse
|
31
|
Valecha GK, Ibrahim U, Ghanem S, Asti D, Atallah JP, Terjanian T. Emerging role of immunotherapy in precursor B-cell acute lymphoblastic leukemia. Expert Rev Hematol 2017; 10:783-799. [DOI: 10.1080/17474086.2017.1350165] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Affiliation(s)
| | - Uroosa Ibrahim
- Department of Hematology-Oncology, Staten Island University Hospital, Staten Island, NY, USA
| | - Sassine Ghanem
- Department of Medicine, Staten Island University Hospital, Staten Island, NY, USA
| | - Divya Asti
- Department of Medicine, Staten Island University Hospital, Staten Island, NY, USA
| | - Jean-Paul Atallah
- Department of Hematology-Oncology, Staten Island University Hospital, Staten Island, NY, USA
| | - Terenig Terjanian
- Department of Hematology-Oncology, Staten Island University Hospital, Staten Island, NY, USA
| |
Collapse
|
32
|
Terwilliger T, Abdul-Hay M. Acute lymphoblastic leukemia: a comprehensive review and 2017 update. Blood Cancer J 2017; 7:e577. [PMID: 28665419 PMCID: PMC5520400 DOI: 10.1038/bcj.2017.53] [Citation(s) in RCA: 745] [Impact Index Per Article: 93.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2017] [Accepted: 04/21/2017] [Indexed: 01/06/2023] Open
Abstract
Acute lymphoblastic leukemia (ALL) is the second most common acute leukemia in adults, with an incidence of over 6500 cases per year in the United States alone. The hallmark of ALL is chromosomal abnormalities and genetic alterations involved in differentiation and proliferation of lymphoid precursor cells. In adults, 75% of cases develop from precursors of the B-cell lineage, with the remainder of cases consisting of malignant T-cell precursors. Traditionally, risk stratification has been based on clinical factors such age, white blood cell count and response to chemotherapy; however, the identification of recurrent genetic alterations has helped refine individual prognosis and guide management. Despite advances in management, the backbone of therapy remains multi-agent chemotherapy with vincristine, corticosteroids and an anthracycline with allogeneic stem cell transplantation for eligible candidates. Elderly patients are often unable to tolerate such regimens and carry a particularly poor prognosis. Here, we review the major recent advances in the treatment of ALL.
Collapse
Affiliation(s)
- T Terwilliger
- New York University School of Medicine, New York, USA
| | - M Abdul-Hay
- New York University School of Medicine, New York, USA
- Department of Hematology, New York University Perlmutter Cancer Center, New York, USA
| |
Collapse
|
33
|
Santiago R, Vairy S, Sinnett D, Krajinovic M, Bittencourt H. Novel therapy for childhood acute lymphoblastic leukemia. Expert Opin Pharmacother 2017; 18:1081-1099. [PMID: 28608730 DOI: 10.1080/14656566.2017.1340938] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/19/2022]
Abstract
INTRODUCTION During recent decades, the prognosis of childhood acute lymphoblastic leukemia (ALL) has improved dramatically, nowadays, reaching a cure rate of almost 90%. These results are due to a better management and combination of old therapies, refined risk-group stratification and emergence of minimal residual disease (MRD) combined with treatment's intensification for high-risk subgroups. However, the subgroup of patients with refractory/relapsed ALL still presents a dismal prognosis indicating necessity for innovative therapeutic approaches. Areas covered: We performed an exhaustive review of current first-line therapies for childhood ALL in the worldwide main consortia, summarized the major advances for front-line and relapse treatment and highlighted recent and promising innovative therapies with an overview of the most promising ongoing clinical trials. Expert opinion: Two major avenues marked the beginning of 21st century. First, is the introduction of tyrosine-kinase inhibitor coupled to chemotherapy for treatment of Philadelphia positive ALL opening new treatment possibilities for the recently identified subgroup of Ph-like ALL. Second, is the breakthrough of immunotherapy, notably CAR T-cell and specific antibody-based therapy, with remarkable success observed in initial studies. This review gives an insight on current knowledge in these innovative therapeutic directions, summarizes currently ongoing clinical trials and addresses challenges these approaches are faced with.
Collapse
Affiliation(s)
- Raoul Santiago
- a CHU Sainte-Justine Research Center , Charles-Bruneau Cancer Center , Montreal , Quebec , Canada.,b Department of Pediatrics, Faculty of Medicine , University of Montreal , Montreal , Quebec , Canada
| | - Stéphanie Vairy
- a CHU Sainte-Justine Research Center , Charles-Bruneau Cancer Center , Montreal , Quebec , Canada.,b Department of Pediatrics, Faculty of Medicine , University of Montreal , Montreal , Quebec , Canada
| | - Daniel Sinnett
- a CHU Sainte-Justine Research Center , Charles-Bruneau Cancer Center , Montreal , Quebec , Canada.,b Department of Pediatrics, Faculty of Medicine , University of Montreal , Montreal , Quebec , Canada
| | - Maja Krajinovic
- a CHU Sainte-Justine Research Center , Charles-Bruneau Cancer Center , Montreal , Quebec , Canada.,b Department of Pediatrics, Faculty of Medicine , University of Montreal , Montreal , Quebec , Canada.,c Department of Pharmacology and Physiology, Faculty of Medicine , University of Montreal , Montreal , Quebec , Canada
| | - Henrique Bittencourt
- a CHU Sainte-Justine Research Center , Charles-Bruneau Cancer Center , Montreal , Quebec , Canada.,b Department of Pediatrics, Faculty of Medicine , University of Montreal , Montreal , Quebec , Canada
| |
Collapse
|
34
|
Chevallier P, Chantepie S, Huguet F, Raffoux E, Thomas X, Leguay T, Marchand T, Isnard F, Charbonnier A, Maury S, Gallego-Hernanz MP, Robillard N, Guillaume T, Peterlin P, Garnier A, Rialland F, Le Houerou C, Goldenberg DM, Wegener WA, Béné MC, Dombret H. Hyper-CVAD + epratuzumab as a salvage regimen for younger patients with relapsed/refractory CD22-positive precursor B-cell acute lymphocytic leukemia. Haematologica 2017; 102:e184-e186. [PMID: 28154086 DOI: 10.3324/haematol.2016.159905] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022] Open
Affiliation(s)
| | | | | | | | | | | | | | | | - Aude Charbonnier
- Hematology Department, Institut Paoli-Calmette, Marseille, France
| | | | | | | | | | | | - Alice Garnier
- Hematology Department, CHU Hotel-Dieu, Nantes, France
| | | | | | | | | | - Marie-C Béné
- Hematology/Biology, CHU Hotel-Dieu, Nantes, France
| | - Hervé Dombret
- Hematology Department, Hôpital Saint-Louis, Paris, France
| |
Collapse
|
35
|
Shah NN. Antibody Based Therapies in Acute Leukemia. Curr Drug Targets 2017; 18:257-270. [PMID: 27593687 PMCID: PMC8335750 DOI: 10.2174/1389450117666160905091459] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2014] [Revised: 03/24/2015] [Accepted: 11/09/2015] [Indexed: 01/05/2023]
Abstract
Despite great progress in the curative treatment of acute leukemia, outcomes for those with relapsed and/or chemotherapy-refractory disease remain poor. Current intensive cytotoxic therapies can be associated with significant morbidity and novel therapies are needed to improve outcomes. Immunotherapy based approaches provide an alternative mechanism of action in the treatment of acute leukemia. Due to cell surface antigen expression, leukemia in particular is amenable to targeted therapies, such as antibody-based therapy. Based on the potential for non-overlapping toxicity, the possibility of synergistic action with standard chemotherapy, and by providing a novel method to overcome chemotherapy resistance, antibody-based therapies have shown potential for benefit. Modifications to standard monoclonal antibodies, including drug conjugation and linkage to T-cells, may further enhance efficacy of antibody-based therapies. Identifying the ideal timing for incorporation of antibody-based therapies, within standard regimens, may lead to improvement in overall outcomes. This article will provide an overview of antibody-based therapies in clinical development for the treatment of acute leukemia in children and adults, with a particular focus on the current strategies and future developments.
Collapse
Affiliation(s)
- Nirali N. Shah
- Pediatric Oncology Branch, Center for Cancer Research (CCR), National Cancer Institute (NCI), NIH, Bethesda, MD, USA
| |
Collapse
|
36
|
Huguet F, Tavitian S. Emerging biological therapies to treat acute lymphoblastic leukemia. Expert Opin Emerg Drugs 2016; 22:107-121. [DOI: 10.1080/14728214.2016.1257606] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023]
|
37
|
Boyiadzis M, Bishop MR, Abonour R, Anderson KC, Ansell SM, Avigan D, Barbarotta L, Barrett AJ, Van Besien K, Bergsagel PL, Borrello I, Brody J, Brufsky J, Cairo M, Chari A, Cohen A, Cortes J, Forman SJ, Friedberg JW, Fuchs EJ, Gore SD, Jagannath S, Kahl BS, Kline J, Kochenderfer JN, Kwak LW, Levy R, de Lima M, Litzow MR, Mahindra A, Miller J, Munshi NC, Orlowski RZ, Pagel JM, Porter DL, Russell SJ, Schwartz K, Shipp MA, Siegel D, Stone RM, Tallman MS, Timmerman JM, Van Rhee F, Waller EK, Welsh A, Werner M, Wiernik PH, Dhodapkar MV. The Society for Immunotherapy of Cancer consensus statement on immunotherapy for the treatment of hematologic malignancies: multiple myeloma, lymphoma, and acute leukemia. J Immunother Cancer 2016; 4:90. [PMID: 28018601 PMCID: PMC5168808 DOI: 10.1186/s40425-016-0188-z] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2016] [Accepted: 11/07/2016] [Indexed: 12/24/2022] Open
Abstract
Increasing knowledge concerning the biology of hematologic malignancies as well as the role of the immune system in the control of these diseases has led to the development and approval of immunotherapies that are resulting in impressive clinical responses. Therefore, the Society for Immunotherapy of Cancer (SITC) convened a hematologic malignancy Cancer Immunotherapy Guidelines panel consisting of physicians, nurses, patient advocates, and patients to develop consensus recommendations for the clinical application of immunotherapy for patients with multiple myeloma, lymphoma, and acute leukemia. These recommendations were developed following the previously established process based on the Institute of Medicine's clinical practice guidelines. In doing so, a systematic literature search was performed for high-impact studies from 2004 to 2014 and was supplemented with further literature as identified by the panel. The consensus panel met in December of 2014 with the goal to generate consensus recommendations for the clinical use of immunotherapy in patients with hematologic malignancies. During this meeting, consensus panel voting along with discussion were used to rate and review the strength of the supporting evidence from the literature search. These consensus recommendations focus on issues related to patient selection, toxicity management, clinical endpoints, and the sequencing or combination of therapies. Overall, immunotherapy is rapidly emerging as an effective therapeutic strategy for the management of hematologic malignances. Evidence-based consensus recommendations for its clinical application are provided and will be updated as the field evolves.
Collapse
Affiliation(s)
- Michael Boyiadzis
- Division of Hematology-Oncology, Department of Medicine, University of Pittsburgh School of Medicine, University of Pittsburgh Cancer Institute, University of Pittsburgh Medical Center, 5150 Centre Avenue, Suite 564, Pittsburg, PA 15232 USA
| | - Michael R. Bishop
- Hematopoietic Cellular Therapy Program, University of Chicago, 5841 S. Maryland Avenue, Chicago, IL 60637 USA
| | - Rafat Abonour
- Indiana University School of Medicine, 980 W. Walnut St., Walther Hall-R3, C400, Indianapolis, IN 46202 USA
| | | | | | - David Avigan
- Beth Israel Deaconess Medical Center, 330 Brookline Avenue, Boston, MA 02215 USA
| | - Lisa Barbarotta
- Smilow Cancer Hospital at Yale New Haven, 35 Park Street, New Haven, CT 06519 USA
| | - Austin John Barrett
- National Institutes of Health, Building 10-CRC Room 3-5330, Bethesda, MD 20814 USA
| | - Koen Van Besien
- Weill Cornell Medical College, 407 E 71st St, New York, NY 10065 USA
| | | | - Ivan Borrello
- Johns Hopkins School of Medicine, 1650 Orleans St, Baltimore, MD 21231 USA
| | - Joshua Brody
- Icahn School of Medicine at Mount Sinai, One Gustave L Levy Place, New York, NY 10029 USA
| | - Jill Brufsky
- University of Pittsburgh Cancer Institute, 5150 Centre Avenue, Pittsburgh, PA 15232 USA
| | - Mitchell Cairo
- New York Medical College at Maria Fareri Children’s Hospital, 100 Woods Road, Valhalla, New York 10595 USA
| | - Ajai Chari
- Icahn School of Medicine at Mount Sinai, One Gustave L Levy Place, New York, NY 10029 USA
| | - Adam Cohen
- Abramson Cancer Center at the University of Pennsylvania, 3400 Civic Center Blvd, Philadelphia, PA 19104 USA
| | - Jorge Cortes
- MD Anderson Cancer Center, 1515 Holcombe Blvd, Houston, TX 77030 USA
| | - Stephen J. Forman
- City of Hope National Medical Center, 1500 East Duarte Road, Duarte, CA 91010 USA
| | - Jonathan W. Friedberg
- Wilmot Cancer Institute, University of Rochester, 601 Elmwood Avenue, Box 704, Rochester, NY 14642 USA
| | - Ephraim J. Fuchs
- Johns Hopkins University School of Medicine, 401 N. Broadway, Baltimore, MD 21231 USA
| | - Steven D. Gore
- Yale Cancer Center, 333 Cedar Street, New Haven, CT 06511 USA
| | - Sundar Jagannath
- Icahn School of Medicine at Mount Sinai, One Gustave L Levy Place, New York, NY 10029 USA
| | - Brad S. Kahl
- Washington University School of Medicine, 660 S. Euclid Ave., St. Louis, MO 63110 USA
| | - Justin Kline
- The University of Chicago, 5841 S. Maryland Ave, Chicago, IL 60637 USA
| | - James N. Kochenderfer
- National Institutes of Health, National Cancer Institute, 8500 Roseweood Drive, Bethesda, MD 20814 USA
| | - Larry W. Kwak
- City of Hope National Medical Center, 1500 E. Duarte Road, Beckman Bldg., Room 4117, Duarte, CA 91010 USA
| | - Ronald Levy
- Division of Medical Oncology, Stanford University School of Medicine, 269 Campus Drive, Stanford, CA 94305 USA
| | - Marcos de Lima
- Department of Medicine-Hematology and Oncology, Case Western Reserve University, 11100 Euclid Ave., Cleveland, OH 44106 USA
| | - Mark R. Litzow
- Department of Hematology, Mayo Clinic Cancer Center, 200 First Street SW, Rochester, MN 55905 USA
| | - Anuj Mahindra
- Helen Diller Family Comprehensive Cancer Center, University of California San Francisco, Box 0324, San Francisco, CA 94143 USA
| | - Jeffrey Miller
- Division of Hematology/Oncology, University of Minnesota, 420 Delaware St SE, Minneapolis, MN 55455 USA
| | - Nikhil C. Munshi
- Dana-Farber Cancer Institute, 450 Brookline Avenue, Dana B106, Boston, MA 02215 USA
| | - Robert Z. Orlowski
- The University of Texas MD Anderson Cancer Center, 1515 Holcombe Blvd., Unit 429, Houston, TX 77030 USA
| | - John M. Pagel
- Swedish Cancer Institute, 1221 Madison Street, Suite 1020, Seattle, WA 98104 USA
| | - David L. Porter
- University of Pennsylvania, 3400 Civic Center Blvd, PCAM 12 South Pavilion, Philadelphia, PA 19104 USA
| | | | - Karl Schwartz
- Patients Against Lymphoma, 3774 Buckwampum Road, Riegelsville, PA 18077 USA
| | - Margaret A. Shipp
- Dana-Farber Cancer Institute, 450 Brookline Ave, Mayer 513, Boston, MA 02215 USA
| | - David Siegel
- Hackensack University Medical Center, 92 2nd St., Hackensack, NJ 07601 USA
| | - Richard M. Stone
- Dana-Farber Cancer Institute, 450 Brookline Avenue, Boston, MA 02215 USA
| | - Martin S. Tallman
- Memorial Sloan Kettering Cancer Center, 1275 York Avenue, New York, NY 10065 USA
| | - John M. Timmerman
- University of California, Los Angeles, 10833 LeConte Ave., Los Angeles, CA 90095 USA
| | - Frits Van Rhee
- University of Arkansas for Medical Sciences, Myeloma Institute, 4301 W Markham #816, Little Rock, AR 72205 USA
| | - Edmund K. Waller
- Winship Cancer Institute, Emory University, 1365B Clifton Road NE, Atlanta, GA 30322 USA
| | - Ann Welsh
- University of Pittsburgh Medical Center, 200 Lothrop St., Pittsburgh, PA 15213 USA
| | - Michael Werner
- Patient Advocate, 33 East Bellevue Place, Chicago, IL 60611 USA
| | - Peter H. Wiernik
- Cancer Research Foundation of New York, 43 Longview Lane, Chappaqua, NY 10514 USA
| | - Madhav V. Dhodapkar
- Department of Hematology & Immunobiology, Yale University, 333 Cedar Street, Box 208021, New Haven, CT 06510 USA
| |
Collapse
|
38
|
Thomas X, Le Jeune C. Treating adults with acute lymphocytic leukemia: new pharmacotherapy options. Expert Opin Pharmacother 2016; 17:2319-2330. [PMID: 27759440 DOI: 10.1080/14656566.2016.1250884] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/20/2022]
Abstract
INTRODUCTION Advances in acute lymphocytic leukemia (ALL) therapy has led to long-term survival rates in children. However, only 30%-40% of adults achieve long-term disease-free survival. After relapse, the outcome of salvage chemotherapy is very disappointing with less than 10% of long survival. Novel agents are therefore desperately required to improve response rates and survival, but also the quality of life of patients. Areas covered: The following review is a comprehensive summary of various novel options reported over the past few years in the therapeutic area of adult ALL. Expert opinion: Identifying key components involved in disease pathogenesis may lead to new approaches. In a near future, the incorporation of monoclonal antibodies and T-cell directed approaches including blinatumomab and chimeric antigen receptor T cells may increase the cure rates and may reduce the need for intensive therapy.
Collapse
Affiliation(s)
- Xavier Thomas
- a Hematology Department , Hospices Civils de Lyon, Lyon-Sud Hospital , Pierre Bénite , France
| | - Caroline Le Jeune
- a Hematology Department , Hospices Civils de Lyon, Lyon-Sud Hospital , Pierre Bénite , France
| |
Collapse
|
39
|
Papadantonakis N, Advani AS. Recent advances and novel treatment paradigms in acute lymphocytic leukemia. Ther Adv Hematol 2016; 7:252-269. [PMID: 27695616 PMCID: PMC5026289 DOI: 10.1177/2040620716652289] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
This is an exciting time in the treatment of acute lymphoblastic leukemia (ALL) given the advances in the relapsed/refractory setting. The development of antibody treatments (including antibody drug conjugates with toxins) offers a different treatment approach compared with conventional chemotherapy regimens. Moreover, the use of bispecific T-cell-engager antibodies (BiTEs) such as blinatumomab harness the cytotoxic activity of T cells against CD19-positive lymphoblasts. Another strategy involves the use of chimeric antigen receptor (CAR) T cells. CAR T cells have demonstrated promising results in the relapsed/refractory setting. However, the use of BiTEs and CAR T cells is also associated with a distinct set of adverse reactions that must be taken into account by the treating physician. Apart from the above strategies, the use of other targeted therapies has attracted interest. Namely, the discovery of the Philadelphia (Ph)-like signature in children and young adults with ALL has led to the use of tyrosine kinase inhibitors (TKI) in these patients. The different drugs and strategies that are being tested in the relapsed/refractory ALL setting pose a unique challenge in identifying the optimum sequence of treatment and determining which approaches should be considered for frontline treatment.
Collapse
Affiliation(s)
| | - Anjali S. Advani
- Taussig Cancer Center, Cleveland Clinic Foundation, Cleveland, OH 44120, USA
| |
Collapse
|
40
|
Monoclonal antibodies and immune therapies for adult precursor B-acute lymphoblastic leukemia. Immunol Lett 2016; 172:113-23. [DOI: 10.1016/j.imlet.2016.02.014] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2016] [Accepted: 02/21/2016] [Indexed: 01/06/2023]
|
41
|
Bodet-Milin C, Kraeber-Bodéré F, Eugène T, Guérard F, Gaschet J, Bailly C, Mougin M, Bourgeois M, Faivre-Chauvet A, Chérel M, Chevallier P. Radioimmunotherapy for Treatment of Acute Leukemia. Semin Nucl Med 2016; 46:135-46. [DOI: 10.1053/j.semnuclmed.2015.10.007] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
|
42
|
Wurz GT, Kao CJ, DeGregorio MW. Novel cancer antigens for personalized immunotherapies: latest evidence and clinical potential. Ther Adv Med Oncol 2016; 8:4-31. [PMID: 26753003 DOI: 10.1177/1758834015615514] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
The clinical success of monoclonal antibody immune checkpoint modulators such as ipilimumab, which targets cytotoxic T lymphocyte-associated antigen 4 (CTLA-4), and the recently approved agents nivolumab and pembrolizumab, which target programmed cell death receptor 1 (PD-1), has stimulated renewed enthusiasm for anticancer immunotherapy, which was heralded by Science as 'Breakthrough of the Year' in 2013. As the potential of cancer immunotherapy has been recognized since the 1890s when William Coley showed that bacterial products could be beneficial in cancer patients, leveraging the immune system in the treatment of cancer is certainly not a new concept; however, earlier attempts to develop effective therapeutic vaccines and antibodies against solid tumors, for example, melanoma, frequently met with failure due in part to self-tolerance and the development of an immunosuppressive tumor microenvironment. Increased knowledge of the mechanisms through which cancer evades the immune system and the identification of tumor-associated antigens (TAAs) and negative immune checkpoint regulators have led to the development of vaccines and monoclonal antibodies targeting specific tumor antigens and immune checkpoints such as CTLA-4 and PD-1. This review first discusses the established targets of currently approved cancer immunotherapies and then focuses on investigational cancer antigens and their clinical potential. Because of the highly heterogeneous nature of tumors, effective anticancer immunotherapy-based treatment regimens will likely require a personalized combination of therapeutic vaccines, antibodies and chemotherapy that fit the specific biology of a patient's disease.
Collapse
Affiliation(s)
- Gregory T Wurz
- Department of Internal Medicine, Division of Hematology and Oncology, University of California, Davis, Sacramento, CA, USA
| | - Chiao-Jung Kao
- Department of Obstetrics and Gynecology, University of California, Davis Sacramento, CA, USA
| | - Michael W DeGregorio
- Department of Internal Medicine, Division of Hematology and Oncology, University of California, Davis, 4501 X Street Suite 3016, Sacramento, CA 95817, USA
| |
Collapse
|
43
|
Chang CH, Wang Y, Gupta P, Goldenberg DM. Extensive crosslinking of CD22 by epratuzumab triggers BCR signaling and caspase-dependent apoptosis in human lymphoma cells. MAbs 2015; 7:199-211. [PMID: 25484043 PMCID: PMC4622945 DOI: 10.4161/19420862.2014.979081] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
Epratuzumab has demonstrated therapeutic activity in patients with non-Hodgkin lymphoma, acute lymphoblastic leukemia, systemic lupus erythematosus, and Sjögren's syndrome, but its mechanism of affecting normal and malignant B cells remains incompletely understood. We reported previously that epratuzumab displayed in vitro cytotoxicity to CD22-expressing Burkitt lymphoma cell lines (Daudi and Ramos) only when immobilized on plates or combined with a crosslinking antibody plus a suboptimal amount of anti-IgM (1 μg/mL). Herein, we show that, in the absence of additional anti-IgM ligation, extensive crosslinking of CD22 by plate-immobilized epratuzumab induced intracellular changes in Daudi cells similar to ligating B-cell antigen receptor with a sufficiently high amount of anti-IgM (10 μg/mL). Specifically, either treatment led to phosphorylation of CD22, CD79a and CD79b, along with their translocation to lipid rafts, both of which were essential for effecting caspase-dependent apoptosis. Moreover, such immobilization induced stabilization of F-actin, phosphorylation of Lyn, ERKs and JNKs, generation of reactive oxygen species (ROS), decrease in mitochondria membrane potential (Δψm), upregulation of pro-apoptotic Bax, and downregulation of anti-apoptotic Bcl-xl and Mcl-1. The physiological relevance of immobilized epratuzumab was implicated by noting that several of its in vitro effects, including apoptosis, drop in Δψm, and generation of ROS, could be observed with soluble epratuzumab in Daudi cells co-cultivated with human umbilical vein endothelial cells. These results suggest that the in vivo mechanism of non-ligand-blocking epratuzumab may, in part, involve the unmasking of CD22 to facilitate the trans-interaction of B cells with vascular endothelium.
Collapse
Key Words
- 488-annexin V, Alexa Fluor 488-conjugated annexin V
- 7-AAD, 7-aminoactinomycin D, Syk, spleen tyrosine kinase
- Anti-IgM, F(ab’)2 fragment of affinity-purified goat anti-human IgM, Fc5μ fragment
- BCR
- BCR, B-cell antigen receptor
- BSA, bovine serum albumin
- CD22
- CM-H2DCF-DA, 2′,7′-dichlorodihydrofluorescein diacetate
- DNP, 2,4-dinitrophenyl
- EC, endothelial cells
- ERKs, extracellular signal-regulated kinases
- FBS, fetal bovine serum
- FITC-DNase I, fluorescein isothiocyanate-conjugated DNase I
- GAH, F(ab′)2 fragment of affinity-purified goat anti-human IgG Fcγ fragment-specific
- HUV-EC
- HUV-EC, human umbilical vein endothelial cells
- ITIM, immunoreceptor tyrosine-based inhibition motif
- JNKs, c-Jun N-terminal kinases
- JP, jasplakinolide
- LatB, latrunculin B
- Lyn, Lck/Yes novel tyrosine kinase
- MAP kinases, mitogen-activated protein kinases
- MTS, (3-(4, 5-dimethylthiazol-2-yl)-5-(3-carboxymethoxyphenyl)-2-(4-sulfophenyl)-2H-tetrazolium
- PARP, poly(ADP-ribose) polymerase
- PBS, phosphate-buffered saline
- PLCγ2, phospholipase C, isotype gamma 2
- ROS, reactive oxygen species
- Rhodamine-anti-IgG, rhodamine-conjugated F(ab′)2 fragment of affinity-purified goat anti-human IgG, F(ab′)2 fragment-specific
- TMRE/tetramethylrhodamine/ethyl ester
- epratuzumab
- human B-cell lymphoma
- immobilized
- mIgM, membrane IgM
- Δψm, mitochondria membrane potential
Collapse
|
44
|
Wolach O, Stone RM. Blinatumomab for the Treatment of Philadelphia Chromosome-Negative, Precursor B-cell Acute Lymphoblastic Leukemia. Clin Cancer Res 2015; 21:4262-9. [PMID: 26283683 DOI: 10.1158/1078-0432.ccr-15-0125] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2015] [Accepted: 07/14/2015] [Indexed: 11/16/2022]
Abstract
Blinatumomab is a CD19/CD3 bispescific antibody designed to redirect T cells toward malignant B cells and induce their lysis. It recently gained accelerated approval by the FDA for the treatment of relapsed or refractory Philadelphia chromosome-negative B-cell acute lymphoblastic leukemia (RR-ALL). In the phase II trial that served as the basis for approval, blinatumomab demonstrated significant single-agent activity and induced remission [complete remission (CR) and CR with incomplete recovery of peripheral blood counts (CRh)] in 43% of 189 adult patients with RR-ALL; the majority of responders (82%) also attained negative minimal residual disease (MRD(-)) status that did not generally translate into long-term remissions in most cases. Additional studies show that blinatumomab can induce high response rates associated with lasting remissions in patients in first remission treated for MRD positivity, suggesting a role for blinatumomab in the upfront, MRD-positive setting. Blinatumomab infusion follows a predictable immunopharmacologic profile, including early cytokine release that can be associated with a clinical syndrome, T-cell expansion, and B-cell depletion. Neurologic toxicities represent a unique toxicity that shares similarities with adverse effects of other T-cell engaging therapies. Further studies are needed to clarify the optimal disease setting and timing for blinatumomab therapy. Additional insights into the pathogenesis, risk factors, and prevention of neurologic toxicities as well as a better understanding of the clinical consequences and biologic pathways that are associated with drug resistance are needed.
Collapse
Affiliation(s)
- Ofir Wolach
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts
| | - Richard M Stone
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts.
| |
Collapse
|
45
|
Raetz EA, Cairo MS, Borowitz MJ, Lu X, Devidas M, Reid JM, Goldenberg DM, Wegener WA, Zeng H, Whitlock JA, Adamson PC, Hunger SP, Carroll WL. Re-induction chemoimmunotherapy with epratuzumab in relapsed acute lymphoblastic leukemia (ALL): Phase II results from Children's Oncology Group (COG) study ADVL04P2. Pediatr Blood Cancer 2015; 62:1171-5. [PMID: 25732247 PMCID: PMC4701208 DOI: 10.1002/pbc.25454] [Citation(s) in RCA: 79] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/11/2014] [Accepted: 01/06/2015] [Indexed: 11/06/2022]
Abstract
BACKGROUND Given the success of immunotherapeutic approaches in hematologic malignancies, the COG designed a phase I/II study to determine whether the addition of epratuzumab (anti-CD22) to an established chemotherapy platform improves rates of second remission (CR2) in pediatric patients with B-lymphoblastic leukemia (B-ALL) and early bone marrow relapse. PROCEDURE Therapy consisted of three established blocks of re-induction chemotherapy. Epratuzumab (360 mg/m(2)/dose) was combined with chemotherapy on weekly × 4 (B1) and twice weekly × 4 [eight doses] (B2) schedules during the first re-induction block. Remission rates and minimal residual disease (MRD) status were compared to historical rates observed with the identical chemotherapy platform alone. RESULTS CR2 was achieved in 65 and 66%, of the evaluable B1 (n = 54) and B2 patients (n = 60), respectively; unchanged from that observed historically without epratuzumab. Rates of MRD negativity (<0.01%) were 31% in B1 (P = 0.4128) and 39% in B2 patients (P = 0.1731), compared to 25% in historical controls. The addition of epratuzumab was well tolerated, with a similar toxicity profile to that observed with the re-induction chemotherapy platform regimen alone. CONCLUSIONS Epratuzumab was well tolerated in combination with re-induction chemotherapy. While CR2 rates were not improved compared to historical controls treated with chemotherapy alone, there was a non-significant trend towards improvement in MRD response with the addition of epratuzumab (twice weekly for eight doses) to re-induction chemotherapy.
Collapse
Affiliation(s)
- Elizabeth A. Raetz
- Department of Pediatrics and Huntsman Cancer Institute, University of Utah, Salt Lake City, UT
| | | | | | - Xiaomin Lu
- Children’s Oncology Group, Gainesville, Florida
| | | | - Joel M. Reid
- Department of Oncology, Mayo Clinic, Rochester, Minnesota
| | | | | | - Hui Zeng
- Children’s Oncology Group, Gainesville, Florida
| | - James A. Whitlock
- Department of Pediatrics, University of Toronto and the Hospital for Sick Children, Toronto, Ontario
| | - Peter C. Adamson
- Department of Pediatrics, Children’s Hospital of Philadelphia, Philadelphia, Pennsylvania
| | - Stephen P. Hunger
- Department of Pediatrics, Children’s Hospital of Philadelphia, Philadelphia, Pennsylvania
| | - William L. Carroll
- NYU Cancer Institute and Department of Pediatrics, NYU Langone Medical Center, New York, New York
| |
Collapse
|
46
|
Bumma N, Papadantonakis N, Advani AS. Structure, development, preclinical and clinical efficacy of blinatumomab in acute lymphoblastic leukemia. Future Oncol 2015; 11:1729-39. [DOI: 10.2217/fon.15.84] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
ABSTRACT The treatment of acute lymphoblastic leukemia (ALL) in adults remains challenging and novel therapies are needed. The antigen, CD19, is expressed by >90% of pre-B ALLs and represents an attractive therapeutic target. The bispecific T-cell-engaging antibody, blinatumomab, targets CD19 and has demonstrated encouraging results in minimal residual disease positive and relapsed/refractory pre-B ALL. In this review, we discuss in detail the mechanism of action and key pharmacologic aspects of blinatumomab. In addition, the preclinical studies, clinical studies and toxicities are summarized.
Collapse
Affiliation(s)
- Naresh Bumma
- Department of Internal Medicine, The Cleveland Clinic, Desk R35, 9500 Euclid Avenue, Cleveland, OH 44195, USA
| | - Nikolaos Papadantonakis
- Department of Hematology/Oncology, Cleveland Clinic Lerner College of Medicine, The Cleveland Clinic, Desk R35, 9500 Euclid Avenue, Cleveland, OH 44195, USA
| | - Anjali S Advani
- Department of Hematology/Oncology, Cleveland Clinic Lerner College of Medicine, The Cleveland Clinic, Desk R35, 9500 Euclid Avenue, Cleveland, OH 44195, USA
| |
Collapse
|
47
|
Monoclonal antibodies in acute lymphoblastic leukemia. Blood 2015; 125:4010-6. [PMID: 25999456 DOI: 10.1182/blood-2014-08-596403] [Citation(s) in RCA: 107] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2014] [Accepted: 12/04/2014] [Indexed: 12/21/2022] Open
Abstract
With modern intensive combination polychemotherapy, the complete response (CR) rate in adults with acute lymphoblastic leukemia (ALL) is 80% to 90%, and the cure rate is 40% to 50%. Hence, there is a need to develop effective salvage therapies and combine novel agents with standard effective chemotherapy. ALL leukemic cells express several surface antigens amenable to target therapies, including CD20, CD22, and CD19. Monoclonal antibodies target these leukemic surface antigens selectively and minimize off-target toxicity. When added to frontline chemotherapy, rituximab, an antibody directed against CD20, increases cure rates of adults with Burkitt leukemia from 40% to 80% and those with pre-B ALL from 35% to 50%. Inotuzumab ozogamicin, a CD22 monoclonal antibody bound to calicheamicin, has resulted in marrow CR rates of 55% and a median survival of 6 to 7 months when given to patients with refractory-relapsed ALL. Blinatumomab, a biallelic T cell engaging the CD3-CD19 monoclonal antibody, also resulted in overall response rates of 40% to 50% and a median survival of 6.5 months in a similar refractory-relapsed population. Other promising monoclonal antibodies targeting CD20 (ofatumumab and obinutuzumab) or CD19 or CD20 and bound to different cytotoxins or immunotoxins are under development. Combined modalities of chemotherapy and the novel monoclonal antibodies are under investigation.
Collapse
|
48
|
How I treat adults with relapsed or refractory Philadelphia chromosome-negative acute lymphoblastic leukemia. Blood 2015; 126:589-96. [PMID: 25966988 DOI: 10.1182/blood-2014-09-551937] [Citation(s) in RCA: 53] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2014] [Accepted: 04/02/2015] [Indexed: 12/17/2022] Open
Abstract
The long-term prognosis of adult patients with relapsed Philadelphia chromosome-negative acute lymphoblastic lymphoma (ALL) is poor. Allogeneic stem cell transplant in second remission is the only curative approach and is the goal when feasible. There is no standard chemotherapy regimen for relapsed disease, although a few agents are approved for use in this setting. The bispecific CD19-directed CD3 T-cell engager, blinatumomab, has recently been granted accelerated approval by the US Food and Drug Administration for relapsed or refractory disease of B-cell lineage. For patients with relapsed T-cell ALL, nelarabine is available. Liposomal vincristine is also approved for relapsed disease. When selecting combination chemotherapy salvage options, evaluation of the prior treatment and timing of relapse informs treatment decisions. Monoclonal and cellular investigational therapies are quite promising and should be explored in the appropriate patient.
Collapse
|
49
|
Future of Therapy in Acute Lymphoblastic Leukemia (ALL)—Potential Role of Immune-Based Therapies. Curr Hematol Malig Rep 2015; 10:76-85. [DOI: 10.1007/s11899-015-0251-8] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/23/2022]
|
50
|
Thomas X. Toward effective targeted therapy for the treatment of adult acute lymphoblastic leukemia. Int J Hematol Oncol 2015. [DOI: 10.2217/ijh.15.1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Affiliation(s)
- Xavier Thomas
- Hospices Civils de Lyon, Hematology Department, Lyon-Sud Hospital, 69495 Pierre Bénite, France
| |
Collapse
|