251
|
Jabbour E, O'Brien S, Konopleva M, Kantarjian H. New insights into the pathophysiology and therapy of adult acute lymphoblastic leukemia. Cancer 2015; 121:2517-2528. [PMID: 25891003 PMCID: PMC11726371 DOI: 10.1002/cncr.29383] [Citation(s) in RCA: 183] [Impact Index Per Article: 18.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2015] [Revised: 02/26/2015] [Accepted: 03/02/2015] [Indexed: 01/13/2023]
Abstract
Significant advances have been made in the last decade toward a better understanding of the disease pathogenesis and the development of novel therapies that target specific subsets of adult acute lymphoblastic leukemia (ALL). Risk-adapted strategies are transforming the disease treatment and prognosis. With current treatment regimens, long-term survival is achieved by approximately 50% of patients with B-cell ALL, 50% to 60% of patients with Philadelphia chromosome-positive ALL, and approximately 80% of patients with Burkitt's leukemia. Genomic profiling in ALL has identified new prognostic markers, new therapeutic targets, and novel ALL subtypes. These may be amenable to future targeted therapies that can further improve outcomes. The early recognition of early precursor T-cell ALL, a distinct pathobiological entity with a poor prognosis, is essential for the development of an effective clinical management strategy. The role of monoclonal antibodies and cytotoxic T-cell therapies continues to be defined. Many of the approaches are currently being evaluated for ALL salvage. Their incorporation into frontline adult ALL therapy, in concomitant or sequential strategies, may increase the cure rates to levels achieved in pediatric ALL and may reduce the need for prolonged intensive and maintenance chemotherapy.
Collapse
Affiliation(s)
- Elias Jabbour
- Department of Leukemia, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Susan O'Brien
- Department of Leukemia, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Marina Konopleva
- Department of Leukemia, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Hagop Kantarjian
- Department of Leukemia, The University of Texas MD Anderson Cancer Center, Houston, Texas
| |
Collapse
|
252
|
Ribera JM, Ferrer A, Ribera J, Genescà E. Profile of blinatumomab and its potential in the treatment of relapsed/refractory acute lymphoblastic leukemia. Onco Targets Ther 2015; 8:1567-74. [PMID: 26170691 PMCID: PMC4485855 DOI: 10.2147/ott.s70524] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
The CD19 marker is expressed on the surface of normal and malignant immature or mature B-cells. On the other hand, immunotherapy involving T-cells is a promising modality of treatment for many neoplastic diseases including leukemias and lymphomas. The CD19/CD3-bispecific T-cell-engaging (BiTE®) monoclonal antibody blinatumomab can transiently engage cytotoxic T-cells to CD19+ target B-cells inducing serial perforin-mediated lysis. In the first clinical trial, blinatumomab showed efficacy in non-Hodgkin’s lymphomas, but the most important trials have been conducted in relapsed/refractory (R/R) acute lymphoblastic leukemia (ALL) and in ALL with minimal residual disease. Encouraging reports on the activity of blinatumomab in R/R Philadelphia chromosome-negative B-cell precursor ALL led to its approval by the US Food and Drug Administration on December 3, 2014 after an accelerated review process. This review focuses on the profile of blinatumomab and its activity in R/R ALL.
Collapse
Affiliation(s)
- Josep-Maria Ribera
- Clinical Hematology Department, ICO-Hospital Germans Trias i Pujol, Josep Carreras Research Institute, Universitat Autònoma de Barcelona, Badalona, Spain
| | - Albert Ferrer
- 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
| | - Eulàlia Genescà
- Clinical Hematology Department, ICO-Hospital Germans Trias i Pujol, Josep Carreras Research Institute, Universitat Autònoma de Barcelona, Badalona, Spain
| |
Collapse
|
253
|
Schreiner J, Thommen DS, Herzig P, Bacac M, Klein C, Roller A, Belousov A, Levitsky V, Savic S, Moersig W, Uhlenbrock F, Heinzelmann-Schwarz VA, Umana P, Pisa P, von Bergwelt-Baildon M, Lardinois D, Müller P, Karanikas V, Zippelius A. Expression of inhibitory receptors on intratumoral T cells modulates the activity of a T cell-bispecific antibody targeting folate receptor. Oncoimmunology 2015; 5:e1062969. [PMID: 27057429 DOI: 10.1080/2162402x.2015.1062969] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2015] [Revised: 06/08/2015] [Accepted: 06/10/2015] [Indexed: 12/18/2022] Open
Abstract
T-cell bispecific antibodies (TCBs) are a novel therapeutic tool designed to selectively recruit T-cells to tumor cells and simultaneously activate them. However, it is currently unknown whether the dysfunctional state of T-cells, embedded into the tumor microenvironment, imprints on the therapeutic activity of TCBs. We performed a comprehensive analysis of activation and effector functions of tumor-infiltrating T-cells (TILs) in different tumor types, upon stimulation by a TCB targeting folate receptor 1 and CD3 (FolR1-TCB). We observed a considerable heterogeneity in T-cell activation, cytokine production and tumor cell killing upon exposure to FolR1-TCB among different FolR1-expressing tumors. Of note, tumors presenting with a high frequency of PD-1hi TILs displayed significantly impaired tumor cell killing and T-cell function. Further characterization of additional T-cell inhibitory receptors revealed that PD-1hi TILs defined a T-cell subset with particularly high levels of multiple inhibitory receptors compared with PD-1int and PD-1neg T-cells. PD-1 blockade could restore cytokine secretion but not cytotoxicity of TILs in a subset of patients with scarce PD-1hi expressing cells; in contrast, patients with abundance of PD-1hi expressing T-cells did not benefit from PD-1 blockade. Our data highlight that FolR1-TCB is a promising novel immunotherapeutic treatment option which is capable of activating intratumoral T-cells in different carcinomas. However, its therapeutic efficacy may be substantially hampered by a pre-existing dysfunctional state of T-cells, reflected by abundance of intratumoral PD-1hi T-cells. These findings present a rationale for combinatorial approaches of TCBs with other therapeutic strategies targeting T-cell dysfunction.
Collapse
Affiliation(s)
- Jens Schreiner
- Laboratory of Cancer Immunology, Department of Biomedicine , Basel, Switzerland
| | - Daniela S Thommen
- Laboratory of Cancer Immunology, Department of Biomedicine, Basel, Switzerland; Department of Medical Oncology, University Hospital Basel, Basel, Switzerland
| | - Petra Herzig
- Laboratory of Cancer Immunology, Department of Biomedicine , Basel, Switzerland
| | - Marina Bacac
- Roche Pharma Research and Early Development, Roche Innovation Center Zurich , Schlieren, Switzerland
| | - Christian Klein
- Roche Pharma Research and Early Development, Roche Innovation Center Zurich , Schlieren, Switzerland
| | - Andreas Roller
- Roche Pharma Research and Early Development, Roche Innovation Center Penzberg, Roche Innovation Center Penzberg , Penzberg, Germany
| | - Anton Belousov
- Roche Pharma Research and Early Development, Roche Innovation Center Penzberg, Roche Innovation Center Penzberg , Penzberg, Germany
| | - Victor Levitsky
- Roche Pharma Research and Early Development, Roche Innovation Center Zurich , Schlieren, Switzerland
| | - Spasenija Savic
- Institute of Pathology, University Hospital Basel , Basel, Switzerland
| | - Wolfgang Moersig
- Department of Surgery, University Hospital Basel , Basel, Switzerland
| | | | | | - Pablo Umana
- Roche Pharma Research and Early Development, Roche Innovation Center Zurich , Schlieren, Switzerland
| | - Pavel Pisa
- Roche Pharma Research and Early Development, Roche Innovation Center Zurich , Schlieren, Switzerland
| | | | - Didier Lardinois
- Department of Surgery, University Hospital Basel , Basel, Switzerland
| | - Philipp Müller
- Laboratory of Cancer Immunology, Department of Biomedicine , Basel, Switzerland
| | - Vaios Karanikas
- Roche Pharma Research and Early Development, Roche Innovation Center Zurich , Schlieren, Switzerland
| | - Alfred Zippelius
- Laboratory of Cancer Immunology, Department of Biomedicine, Basel, Switzerland; Department of Medical Oncology, University Hospital Basel, Basel, Switzerland
| |
Collapse
|
254
|
Turini M, Chames P, Bruhns P, Baty D, Kerfelec B. A FcγRIII-engaging bispecific antibody expands the range of HER2-expressing breast tumors eligible to antibody therapy. Oncotarget 2015; 5:5304-19. [PMID: 24979648 PMCID: PMC4170649 DOI: 10.18632/oncotarget.2093] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022] Open
Abstract
Trastuzumab is established as treatment of HER2high metastatic breast cancers but many limitations impair its efficacy. Here, we report the design of a Fab-like bispecific antibody (HER2bsFab) that displays a moderate affinity for HER2 and a unique, specific and high affinity for FcγRIII. In vitro characterization showed that ADCC was the major mechanism of action of HER2bsFab as no significant HER2-driven effect was observed. HER2bsFab mediated ADCC at picomolar concentration against HER2high, HER2low as well as trastuzumab-refractive cell lines. In vivo HER2bsFab potently inhibited HER2high tumor growth by recruitment of mouse FcγRIII and IV-positive resident effector cells and more importantly, exhibited a net superiority over trastuzumab at inhibiting HER2low tumor growth. Moreover, FcγRIIIA-engagement by HER2bsFab was independent of V/F158 polymorphism and induced a stronger NK cells activation in response to target cell recognition. Thus, taking advantage of its epitope specificity and affinity for HER2 and FcγRIIIA, HER2bsFab exhibits potent anti-tumor activity against HER2low tumors while evading most of trastuzumab Fc-linked limitations thereby potentially enlarging the number of patients eligible for breast cancer immunotherapy.
Collapse
Affiliation(s)
- Marc Turini
- INSERM, U1068, CRCM, Marseille, France. Institut Paoli-Calmettes, Marseille, France. Aix-Marseille Université, UM105, Marseille, France. CNRS, UMR7258, CRCM, Marseille, France
| | - Patrick Chames
- INSERM, U1068, CRCM, Marseille, France. Institut Paoli-Calmettes, Marseille, France. Aix-Marseille Université, UM105, Marseille, France. CNRS, UMR7258, CRCM, Marseille, France
| | - Pierre Bruhns
- Département d'Immunologie, Laboratoire Anticorps en Thérapie et Pathologie, Institut Pasteur, Paris, France. INSERM, U760, Paris, France
| | - Daniel Baty
- INSERM, U1068, CRCM, Marseille, France. Institut Paoli-Calmettes, Marseille, France. Aix-Marseille Université, UM105, Marseille, France. CNRS, UMR7258, CRCM, Marseille, France
| | - Brigitte Kerfelec
- INSERM, U1068, CRCM, Marseille, France. Institut Paoli-Calmettes, Marseille, France. Aix-Marseille Université, UM105, Marseille, France. CNRS, UMR7258, CRCM, Marseille, France
| |
Collapse
|
255
|
Abstract
The therapeutic potential of the immune system in the context of hematologic malignancies has long been appreciated particularly due to the curative impact of allogeneic hematopoietic stem cell transplantation (SCT). The role of immune system in shaping the biology and evolution of these tumors is now well recognized. While the contribution of the immune system in anti-tumor effects of certain therapies such as immune-modulatory drugs and monoclonal antibodies active in hematologic malignancies is quite evident, the immune system has also been implicated in anti-tumor effects of other targeted therapies. The horizon of immune-based therapies in hematologic malignancies is rapidly expanding with promising results from immune-modulatory drugs, immune-checkpoint blockade, and adoptive cellular therapies, including genetically-modified T cells. Hematologic malignancies present distinct issues (relative to solid tumors) for the application of immune therapies due to differences in cell of origin/developmental niche of tumor cells, and patterns of involvement such as common systemic involvement of secondary lymphoid tissues. This article discusses the rapidly changing landscape of immune modulation in hematologic malignancies and emphasizes areas wherein hematologic malignancies present distinct opportunities for immunologic approaches to prevent or treat cancer.
Collapse
Affiliation(s)
- Madhav V Dhodapkar
- Departments of Internal Medicine (Hematology); Immunobiology, Yale University, New Haven, CT; Yale Cancer Center, Yale University, New Haven, CT.
| | - Kavita M Dhodapkar
- Pediatrics (Hematology-Oncology); Yale Cancer Center, Yale University, New Haven, CT
| |
Collapse
|
256
|
Abstract
Objective: To review the clinical pharmacology, efficacy, and safety of blinatumomab for the treatment of pediatric and adult precursor B-cell acute lymphoblastic leukemia (B-ALL). Data Sources: A literature search of EMBASE (1947 to April 2015), Medline (1946 to April 2015), PubMed (1996 to April 2015), the U.S. National Institutes of Health Clinicaltrials.gov, the Food and Drug Administration, and relevant meeting abstracts was conducted using the terms blinatumomab, BiTE, bispecific T-cell engager, MT103, MEDI-538, and Blincyto. Study Selection/Data Extraction: Human and animal studies describing the pharmacology, pharmacokinetics and pharmacodynamics, efficacy, and safety of blinatumomab for precursor B-ALL were identified. Data Synthesis: Blinatumomab is a first-in-class bispecific T-cell engager (BiTE) antibody derived from a B-lineage specific antitumor mouse monoclonal antibody that binds to both CD19 of B-cells and CD3 of T-cells. A pivotal phase II trial demonstrated that response rates were high in a refractory or relapsed patient population, with 43% achieving complete remission (CR). Median relapse-free survival was 5.9 months for those with CR or CR with incomplete hematological recovery. Median overall survival was 6.1 months, and 60% of patients achieved minimal residual disease (MRD) negativity. The most common adverse events included pyrexia, neurological events, headache, febrile neutropenia, peripheral edema, nausea, hypokalemia, constipation, and anemia. Conclusions: Blinatumomab is a novel BiTE therapeutic monoclonal antibody that has shown promising results in patients with relapsed or refractory ALL or those achieving a CR with persistent MRD. Phase III clinical trials should define the optimal place in therapy of blinatumomab.
Collapse
Affiliation(s)
- Larry W. Buie
- Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | | | - Troy Z. Horvat
- Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Ryan J. Daley
- Memorial Sloan Kettering Cancer Center, New York, NY, USA
| |
Collapse
|
257
|
Osada T, Patel SP, Hammond SA, Osada K, Morse MA, Lyerly HK. CEA/CD3-bispecific T cell-engaging (BiTE) antibody-mediated T lymphocyte cytotoxicity maximized by inhibition of both PD1 and PD-L1. Cancer Immunol Immunother 2015; 64:677-88. [PMID: 25742933 PMCID: PMC11029757 DOI: 10.1007/s00262-015-1671-y] [Citation(s) in RCA: 67] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2014] [Accepted: 02/17/2015] [Indexed: 12/22/2022]
Abstract
Bispecific T cell-engaging (BiTE) antibodies recruit polyclonal cytotoxic T cells (CTL) to tumors. One such antibody is carcinoembryonic antigen (CEA) BiTE that mediates T cell/tumor interaction by simultaneously binding CD3 expressed by T cells and CEA expressed by tumor cells. A widely operative mechanism for mitigating cytotoxic T cell-mediated killing is the interaction of tumor-expressed PD-L1 with T cell-expressed PD-1, which may be partly reversed by PD-1/PD-L1 blockade. We hypothesized that PD-1/PD-L1 blockade during BiTE-mediated T cell killing would enhance CTL function. Here, we determined the effects of PD-1 and PD-L1 blockade during initial T cell-mediated killing of CEA-expressing human tumor cell lines in vitro, as well as subsequent T cell-mediated killing by T lymphocytes that had participated in tumor cell killing. We observed a rapid upregulation of PD-1 expression and diminished cytolytic function of T cells after they had engaged in CEA BiTE-mediated killing of tumors. T cell cytolytic activity in vitro could be maximized by administration of anti-PD-1 or anti-PD-L1 antibodies alone or in combination if applied prior to a round of T cell killing, but T cell inhibition could not be fully reversed by this blockade once the T cells had killed tumor. In conclusion, our findings demonstrate that dual blockade of PD-1 and PD-L1 maximizes T cell killing of tumor directed by CEA BiTE in vitro, is more effective if applied early, and provides a rationale for clinical use.
Collapse
Affiliation(s)
- Takuya Osada
- Section of Applied Therapeutics, Department of Surgery, Duke University Medical Center, 403 MSRB, Research Drive, Durham, NC, 27710, USA,
| | | | | | | | | | | |
Collapse
|
258
|
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
|
259
|
Rogala B, Freyer CW, Ontiveros EP, Griffiths EA, Wang ES, Wetzler M. Blinatumomab: enlisting serial killer T-cells in the war against hematologic malignancies. Expert Opin Biol Ther 2015; 15:895-908. [PMID: 25985814 PMCID: PMC4994468 DOI: 10.1517/14712598.2015.1041912] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023]
Abstract
INTRODUCTION The approval of blinatumomab signals the long awaited arrival of immunotherapy for acute lymphoblastic leukemia (ALL). Previous options for relapsed or refractory disease were restricted to cytotoxic chemotherapy with limited efficacy and significant toxicity. Through an innovative mechanism of action, blinatumomab stimulates a polyclonal antitumor T-cell response, yielding unprecedented single agent efficacy in the relapsed/refractory setting. Success comes at the cost of immunological toxicities rarely encountered with previous therapies and challenging administration logistics requiring clinical expertise. AREAS COVERED All published clinical and preclinical studies using blinatumomab were reviewed in addition to all registered ongoing clinical trials and data published in abstract form. The search was limited to the English language. The pharmacology, clinical efficacy, toxicity profile, and logistical considerations for drug administration are discussed. EXPERT OPINION Blinatumomab is an exciting addition to the treatment armamentarium for relapsed/refractory ALL, yet several questions remain regarding optimal implementation into the current treatment paradigm. A unique toxicity profile should be weighed against promising benefits in a poor prognosis population. Other emerging therapies, such as chimeric antigen receptor-modified T-cells and inotuzumab ozogamicin, with different side effect profiles and administration schedules, may prove to be more beneficial for specific patient populations.
Collapse
Affiliation(s)
- Britny Rogala
- Roswell Park Cancer Institute, Department of Pharmacy. Elm & Carlton Streets, Buffalo, NY, USA 14263
| | - Craig W. Freyer
- Roswell Park Cancer Institute, Department of Pharmacy. Elm & Carlton Streets, Buffalo, NY, USA 14263
| | - Evelena P. Ontiveros
- Roswell Park Cancer Institute, Department of Medicine, Leukemia Section. Elm & Carlton Streets, Buffalo, NY, USA 14263
| | - Elizabeth A. Griffiths
- Roswell Park Cancer Institute, Department of Medicine, Leukemia Section. Elm & Carlton Streets, Buffalo, NY, USA 14263
| | - Eunice S. Wang
- Roswell Park Cancer Institute, Department of Medicine, Leukemia Section. Elm & Carlton Streets, Buffalo, NY, USA 14263
| | - Meir Wetzler
- Roswell Park Cancer Institute, Department of Medicine, Leukemia Section. Elm & Carlton Streets, Buffalo, NY, USA 14263
| |
Collapse
|
260
|
Ruella M, Gill S. How to train your T cell: genetically engineered chimeric antigen receptor T cells versus bispecific T-cell engagers to target CD19 in B acute lymphoblastic leukemia. Expert Opin Biol Ther 2015; 15:761-6. [PMID: 25640460 DOI: 10.1517/14712598.2015.1009888] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
Abstract
Antigen-specific T cell-based immunotherapy is getting its day in the sun. The contemporaneous development of two potent CD19-specific immunotherapeutic modalities for the treatment of B-cell malignancies provides exciting opportunities for patients, physicians and scientists alike. Patients with relapsed, refractory or poor-risk B-cell acute lymphoblastic leukemia (ALL) previously had few therapeutic options and now have two potential new lifelines. Physicians will have the choice between two powerful modalities and indeed could potentially enroll some patients on trials exploring both modalities if needed. For scientists interested in tumor immunology, the advent of chimeric antigen receptor T-cell therapy and of bispecific T-cell engagers (BiTEs) provides unprecedented opportunities to explore the promise and limitations of antigen-specific T-cell therapy in the context of human leukemia. In this article, we compare chimeric antigen receptor T cells and BiTEs targeting CD19 in B-cell ALL in the setting of the available clinical literature.
Collapse
Affiliation(s)
- Marco Ruella
- University of Pennsylvania, Perelman School of Medicine, Translational Research Program , Philadelphia, PA , USA
| | | |
Collapse
|
261
|
Minimal residual disease diagnostics in acute lymphoblastic leukemia: need for sensitive, fast, and standardized technologies. Blood 2015; 125:3996-4009. [PMID: 25999452 DOI: 10.1182/blood-2015-03-580027] [Citation(s) in RCA: 352] [Impact Index Per Article: 35.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2015] [Accepted: 05/10/2015] [Indexed: 12/26/2022] Open
Abstract
Monitoring of minimal residual disease (MRD) has become routine clinical practice in frontline treatment of virtually all childhood acute lymphoblastic leukemia (ALL) and in many adult ALL patients. MRD diagnostics has proven to be the strongest prognostic factor, allowing for risk group assignment into different treatment arms, ranging from significant treatment reduction to mild or strong intensification. Also in relapsed ALL patients and patients undergoing stem cell transplantation, MRD diagnostics is guiding treatment decisions. This is also why the efficacy of innovative drugs, such as antibodies and small molecules, are currently being evaluated with MRD diagnostics within clinical trials. In fact, MRD measurements might well be used as a surrogate end point, thereby significantly shortening the follow-up. The MRD techniques need to be sensitive (≤10(-4)), broadly applicable, accurate, reliable, fast, and affordable. Thus far, flow cytometry and polymerase chain reaction (PCR) analysis of rearranged immunoglobulin and T-cell receptor genes (allele-specific oligonucleotide [ASO]-PCR) are claimed to meet these criteria, but classical flow cytometry does not reach a solid 10(-4), whereas classical ASO-PCR is time-consuming and labor intensive. Therefore, 2 high-throughput technologies are being explored, ie, high-throughput sequencing and next-generation (multidimensional) flow cytometry, both evaluating millions of sequences or cells, respectively. Each of them has specific advantages and disadvantages.
Collapse
|
262
|
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
|
263
|
Leukemic progenitor cells are susceptible to targeting by stimulated cytotoxic T cells against immunogenic leukemia-associated antigens. Int J Cancer 2015; 137:2083-92. [DOI: 10.1002/ijc.29583] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2014] [Accepted: 03/30/2015] [Indexed: 12/30/2022]
|
264
|
Stieglmaier J, Benjamin J, Nagorsen D. Utilizing the BiTE (bispecific T-cell engager) platform for immunotherapy of cancer. Expert Opin Biol Ther 2015; 15:1093-9. [PMID: 25971805 DOI: 10.1517/14712598.2015.1041373] [Citation(s) in RCA: 50] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
Abstract
Various approaches of T-cell-based cancer immunotherapy are currently under investigation, among these are BiTE® (bispecific T-cell engager) antibody constructs, which have a unique design and mechanism of action. They are constructed by genetically linking onto a single polypeptide chain the minimal binding domains of monoclonal antibodies for tumor-associated surface antigens and for the T-cell receptor-associated molecule CD3. Concurrent engagement of the target cell antigen and CD3 leads to activation of polyclonal cytotoxic T-cells, resulting in target cell lysis. Blinatumomab, a BiTE targeting CD19, is being investigated in a broad range of B-cell malignancies and has recently been approved in the USA by the US FDA for Philadelphia chromosome-negative relapsed/refractory B-acute lymphoblastic leukemia under the trade name BLINCYTO™. The BiTE platform is one of the clinically most advanced T-cell immunotherapy options.
Collapse
Affiliation(s)
- Julia Stieglmaier
- Amgen Research (Munich), Global Clinical Development, Therapeutic Area Oncology , Staffelseestraße 2, Munich 81477 , Germany
| | | | | |
Collapse
|
265
|
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
|
266
|
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]
|
267
|
Randomized study of reduced-intensity chemotherapy combined with imatinib in adults with Ph-positive acute lymphoblastic leukemia. Blood 2015; 125:3711-9. [PMID: 25878120 DOI: 10.1182/blood-2015-02-627935] [Citation(s) in RCA: 266] [Impact Index Per Article: 26.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2015] [Accepted: 04/11/2015] [Indexed: 01/28/2023] Open
Abstract
In this study, we randomly compared high doses of the tyrosine kinase inhibitor imatinib combined with reduced-intensity chemotherapy (arm A) to standard imatinib/hyperCVAD (cyclophosphamide/vincristine/doxorubicin/dexamethasone) therapy (arm B) in 268 adults (median age, 47 years) with Philadelphia chromosome-positive (Ph+) acute lymphoblastic leukemia (ALL). The primary objective was the major molecular response (MMolR) rate after cycle 2, patients being then eligible for allogeneic stem cell transplantation (SCT) if they had a donor, or autologous SCT if in MMolR and no donor. With fewer induction deaths, the complete remission (CR) rate was higher in arm A than in arm B (98% vs 91%; P = .006), whereas the MMolR rate was similar in both arms (66% vs 64%). With a median follow-up of 4.8 years, 5-year event-free survival and overall survival (OS) rates were estimated at 37.1% and 45.6%, respectively, without difference between the arms. Allogeneic transplantation was associated with a significant benefit in relapse-free survival (hazard ratio [HR], 0.69; P = .036) and OS (HR, 0.64; P = .02), with initial white blood cell count being the only factor significantly interacting with this SCT effect. In patients achieving MMolR, outcome was similar after autologous and allogeneic transplantation. This study validates an induction regimen combining reduced-intensity chemotherapy and imatinib in Ph+ ALL adult patients and suggests that SCT in first CR is still a good option for Ph+ ALL adult patients. This trial was registered at www.clinicaltrials.gov as #NCT00327678.
Collapse
|
268
|
Zugmaier G, Klinger M, Schmidt M, Subklewe M. Clinical overview of anti-CD19 BiTE(®) and ex vivo data from anti-CD33 BiTE(®) as examples for retargeting T cells in hematologic malignancies. Mol Immunol 2015; 67:58-66. [PMID: 25883042 DOI: 10.1016/j.molimm.2015.02.033] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2014] [Revised: 02/20/2015] [Accepted: 02/28/2015] [Indexed: 02/01/2023]
Abstract
Blinatumomab, a bispecific antibody construct targeting CD19, is the most advanced member of bispecific T-cell engager (BiTE(®)) molecules. The clinical development program includes B-precursor acute lymphoblastic leukemia (ALL) and B-cell non-Hodgkin lymphoma (NHL). Minimal residual disease (MRD) response in patients with MRD-positive B-precursor ALL has translated into long-term clinical benefits as demonstrated by an estimated relapse-free survival (RFS) of 60% with sustained MRD negativity at a follow-up of 31 months. Remissions induced in pediatric and adult patients with relapsed/refractory B-precursor ALL have allowed for successful allogeneic hematopoietic stem cell transplantation (HSCT) in this setting. Blinatumomab has also induced durable responses in low-grade B-cell NHL. Blinatumomab recently gained approval in the United States by the U.S. Food and Drug Administration for treatment of Philadelphia chromosome-negative B-precursor relapsed/refractory acute lymphoblastic leukemia. AMG 330 is an investigational anti-CD33 BiTE(®) antibody construct. Targeting CD33 ex vivo in primary samples from patients with acute myeloid leukemia (AML) has shown AMG 330-mediated T-cell expansion and T-cell cytotoxicity against AML cells.
Collapse
Affiliation(s)
| | | | | | - Marion Subklewe
- Department of Internal Medicine III, Klinikum der Universität München, Munich, Germany; Clinical Cooperation Group Immunotherapy, Helmholtz Institute Munich, Munich, Germany
| |
Collapse
|
269
|
Bader P, Kreyenberg H, von Stackelberg A, Eckert C, Salzmann-Manrique E, Meisel R, Poetschger U, Stachel D, Schrappe M, Alten J, Schrauder A, Schulz A, Lang P, Müller I, Albert MH, Willasch AM, Klingebiel TE, Peters C. Monitoring of Minimal Residual Disease After Allogeneic Stem-Cell Transplantation in Relapsed Childhood Acute Lymphoblastic Leukemia Allows for the Identification of Impending Relapse: Results of the ALL-BFM-SCT 2003 Trial. J Clin Oncol 2015; 33:1275-84. [DOI: 10.1200/jco.2014.58.4631] [Citation(s) in RCA: 101] [Impact Index Per Article: 10.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Purpose To elucidate the impact of minimal residual disease (MRD) after allogeneic transplantation, the Acute Lymphoblastic Leukemia Berlin-Frankfurt-Münster Stem Cell Transplantation Group (ALL-BFM-SCT) conducted a prospective clinical trial. Patients and Methods In the ALL-BFM-SCT 2003 trial, MRD was assessed in the bone marrow at days +30, +60, +90, +180, and +365 after transplantation in 113 patients with relapsed disease. Standardized quantification of MRD was performed according to the guidelines of the Euro-MRD Group. Results All patients showed a 3-year probability of event-free survival (pEFS) of 55%. The cumulative incidence rates of relapse and treatment-related mortality were 32% and 12%, respectively. The pEFS was 60% for patients who received their transplantations in second complete remission, 50% for patients in ≥ third complete remission, and 0% for patients not in remission (P = .015). At all time points, the level of MRD was inversely correlated with event-free survival (EFS; P < .004) and positively correlated with the cumulative incidence of relapse (P < .01). A multivariable Cox model was fitted for each time point, which showed that MRD ≥ 10−4 leukemic cells was consistently correlated with inferior EFS (P < .003). The accuracy of MRD measurements in predicting relapse was investigated with time-dependent receiver operating curves at days +30, +60, +90, and +180. From day +60 onward, the discriminatory power of MRD detection to predict the probability of relapse after 1, 3, 6, and 9 months was more than 96%, more than 87%, more than 71%, and more than 61%, respectively. Conclusion MRD after transplantation was a reliable marker for predicting impending relapses and could thus serve as the basis for pre-emptive therapy.
Collapse
Affiliation(s)
- Peter Bader
- Peter Bader, Hermann Kreyenberg, Emilia Salzmann-Manrique, Andre M. Willasch, and Thomas E. Klingebiel, University Hospital for Children and Adolescents, Frankfurt/Main; Arend von Stackelberg and Cornelia Eckert, Children's Hospital Charité, Berlin, Berlin; Roland Meisel, Heinrich-Heine-University, Düsseldorf; Daniel Stachel, University Hospital Erlangen, Erlangen; Martin Schrappe, Julia Alten, Andre Schrauder, Christian-Albrechts-University and Medical Center Schleswig-Holstein, Kiel; Ansgar Schulz,
| | - Hermann Kreyenberg
- Peter Bader, Hermann Kreyenberg, Emilia Salzmann-Manrique, Andre M. Willasch, and Thomas E. Klingebiel, University Hospital for Children and Adolescents, Frankfurt/Main; Arend von Stackelberg and Cornelia Eckert, Children's Hospital Charité, Berlin, Berlin; Roland Meisel, Heinrich-Heine-University, Düsseldorf; Daniel Stachel, University Hospital Erlangen, Erlangen; Martin Schrappe, Julia Alten, Andre Schrauder, Christian-Albrechts-University and Medical Center Schleswig-Holstein, Kiel; Ansgar Schulz,
| | - Arend von Stackelberg
- Peter Bader, Hermann Kreyenberg, Emilia Salzmann-Manrique, Andre M. Willasch, and Thomas E. Klingebiel, University Hospital for Children and Adolescents, Frankfurt/Main; Arend von Stackelberg and Cornelia Eckert, Children's Hospital Charité, Berlin, Berlin; Roland Meisel, Heinrich-Heine-University, Düsseldorf; Daniel Stachel, University Hospital Erlangen, Erlangen; Martin Schrappe, Julia Alten, Andre Schrauder, Christian-Albrechts-University and Medical Center Schleswig-Holstein, Kiel; Ansgar Schulz,
| | - Cornelia Eckert
- Peter Bader, Hermann Kreyenberg, Emilia Salzmann-Manrique, Andre M. Willasch, and Thomas E. Klingebiel, University Hospital for Children and Adolescents, Frankfurt/Main; Arend von Stackelberg and Cornelia Eckert, Children's Hospital Charité, Berlin, Berlin; Roland Meisel, Heinrich-Heine-University, Düsseldorf; Daniel Stachel, University Hospital Erlangen, Erlangen; Martin Schrappe, Julia Alten, Andre Schrauder, Christian-Albrechts-University and Medical Center Schleswig-Holstein, Kiel; Ansgar Schulz,
| | - Emilia Salzmann-Manrique
- Peter Bader, Hermann Kreyenberg, Emilia Salzmann-Manrique, Andre M. Willasch, and Thomas E. Klingebiel, University Hospital for Children and Adolescents, Frankfurt/Main; Arend von Stackelberg and Cornelia Eckert, Children's Hospital Charité, Berlin, Berlin; Roland Meisel, Heinrich-Heine-University, Düsseldorf; Daniel Stachel, University Hospital Erlangen, Erlangen; Martin Schrappe, Julia Alten, Andre Schrauder, Christian-Albrechts-University and Medical Center Schleswig-Holstein, Kiel; Ansgar Schulz,
| | - Roland Meisel
- Peter Bader, Hermann Kreyenberg, Emilia Salzmann-Manrique, Andre M. Willasch, and Thomas E. Klingebiel, University Hospital for Children and Adolescents, Frankfurt/Main; Arend von Stackelberg and Cornelia Eckert, Children's Hospital Charité, Berlin, Berlin; Roland Meisel, Heinrich-Heine-University, Düsseldorf; Daniel Stachel, University Hospital Erlangen, Erlangen; Martin Schrappe, Julia Alten, Andre Schrauder, Christian-Albrechts-University and Medical Center Schleswig-Holstein, Kiel; Ansgar Schulz,
| | - Ulrike Poetschger
- Peter Bader, Hermann Kreyenberg, Emilia Salzmann-Manrique, Andre M. Willasch, and Thomas E. Klingebiel, University Hospital for Children and Adolescents, Frankfurt/Main; Arend von Stackelberg and Cornelia Eckert, Children's Hospital Charité, Berlin, Berlin; Roland Meisel, Heinrich-Heine-University, Düsseldorf; Daniel Stachel, University Hospital Erlangen, Erlangen; Martin Schrappe, Julia Alten, Andre Schrauder, Christian-Albrechts-University and Medical Center Schleswig-Holstein, Kiel; Ansgar Schulz,
| | - Daniel Stachel
- Peter Bader, Hermann Kreyenberg, Emilia Salzmann-Manrique, Andre M. Willasch, and Thomas E. Klingebiel, University Hospital for Children and Adolescents, Frankfurt/Main; Arend von Stackelberg and Cornelia Eckert, Children's Hospital Charité, Berlin, Berlin; Roland Meisel, Heinrich-Heine-University, Düsseldorf; Daniel Stachel, University Hospital Erlangen, Erlangen; Martin Schrappe, Julia Alten, Andre Schrauder, Christian-Albrechts-University and Medical Center Schleswig-Holstein, Kiel; Ansgar Schulz,
| | - Martin Schrappe
- Peter Bader, Hermann Kreyenberg, Emilia Salzmann-Manrique, Andre M. Willasch, and Thomas E. Klingebiel, University Hospital for Children and Adolescents, Frankfurt/Main; Arend von Stackelberg and Cornelia Eckert, Children's Hospital Charité, Berlin, Berlin; Roland Meisel, Heinrich-Heine-University, Düsseldorf; Daniel Stachel, University Hospital Erlangen, Erlangen; Martin Schrappe, Julia Alten, Andre Schrauder, Christian-Albrechts-University and Medical Center Schleswig-Holstein, Kiel; Ansgar Schulz,
| | - Julia Alten
- Peter Bader, Hermann Kreyenberg, Emilia Salzmann-Manrique, Andre M. Willasch, and Thomas E. Klingebiel, University Hospital for Children and Adolescents, Frankfurt/Main; Arend von Stackelberg and Cornelia Eckert, Children's Hospital Charité, Berlin, Berlin; Roland Meisel, Heinrich-Heine-University, Düsseldorf; Daniel Stachel, University Hospital Erlangen, Erlangen; Martin Schrappe, Julia Alten, Andre Schrauder, Christian-Albrechts-University and Medical Center Schleswig-Holstein, Kiel; Ansgar Schulz,
| | - Andre Schrauder
- Peter Bader, Hermann Kreyenberg, Emilia Salzmann-Manrique, Andre M. Willasch, and Thomas E. Klingebiel, University Hospital for Children and Adolescents, Frankfurt/Main; Arend von Stackelberg and Cornelia Eckert, Children's Hospital Charité, Berlin, Berlin; Roland Meisel, Heinrich-Heine-University, Düsseldorf; Daniel Stachel, University Hospital Erlangen, Erlangen; Martin Schrappe, Julia Alten, Andre Schrauder, Christian-Albrechts-University and Medical Center Schleswig-Holstein, Kiel; Ansgar Schulz,
| | - Ansgar Schulz
- Peter Bader, Hermann Kreyenberg, Emilia Salzmann-Manrique, Andre M. Willasch, and Thomas E. Klingebiel, University Hospital for Children and Adolescents, Frankfurt/Main; Arend von Stackelberg and Cornelia Eckert, Children's Hospital Charité, Berlin, Berlin; Roland Meisel, Heinrich-Heine-University, Düsseldorf; Daniel Stachel, University Hospital Erlangen, Erlangen; Martin Schrappe, Julia Alten, Andre Schrauder, Christian-Albrechts-University and Medical Center Schleswig-Holstein, Kiel; Ansgar Schulz,
| | - Peter Lang
- Peter Bader, Hermann Kreyenberg, Emilia Salzmann-Manrique, Andre M. Willasch, and Thomas E. Klingebiel, University Hospital for Children and Adolescents, Frankfurt/Main; Arend von Stackelberg and Cornelia Eckert, Children's Hospital Charité, Berlin, Berlin; Roland Meisel, Heinrich-Heine-University, Düsseldorf; Daniel Stachel, University Hospital Erlangen, Erlangen; Martin Schrappe, Julia Alten, Andre Schrauder, Christian-Albrechts-University and Medical Center Schleswig-Holstein, Kiel; Ansgar Schulz,
| | - Ingo Müller
- Peter Bader, Hermann Kreyenberg, Emilia Salzmann-Manrique, Andre M. Willasch, and Thomas E. Klingebiel, University Hospital for Children and Adolescents, Frankfurt/Main; Arend von Stackelberg and Cornelia Eckert, Children's Hospital Charité, Berlin, Berlin; Roland Meisel, Heinrich-Heine-University, Düsseldorf; Daniel Stachel, University Hospital Erlangen, Erlangen; Martin Schrappe, Julia Alten, Andre Schrauder, Christian-Albrechts-University and Medical Center Schleswig-Holstein, Kiel; Ansgar Schulz,
| | - Michael H. Albert
- Peter Bader, Hermann Kreyenberg, Emilia Salzmann-Manrique, Andre M. Willasch, and Thomas E. Klingebiel, University Hospital for Children and Adolescents, Frankfurt/Main; Arend von Stackelberg and Cornelia Eckert, Children's Hospital Charité, Berlin, Berlin; Roland Meisel, Heinrich-Heine-University, Düsseldorf; Daniel Stachel, University Hospital Erlangen, Erlangen; Martin Schrappe, Julia Alten, Andre Schrauder, Christian-Albrechts-University and Medical Center Schleswig-Holstein, Kiel; Ansgar Schulz,
| | - Andre M. Willasch
- Peter Bader, Hermann Kreyenberg, Emilia Salzmann-Manrique, Andre M. Willasch, and Thomas E. Klingebiel, University Hospital for Children and Adolescents, Frankfurt/Main; Arend von Stackelberg and Cornelia Eckert, Children's Hospital Charité, Berlin, Berlin; Roland Meisel, Heinrich-Heine-University, Düsseldorf; Daniel Stachel, University Hospital Erlangen, Erlangen; Martin Schrappe, Julia Alten, Andre Schrauder, Christian-Albrechts-University and Medical Center Schleswig-Holstein, Kiel; Ansgar Schulz,
| | - Thomas E. Klingebiel
- Peter Bader, Hermann Kreyenberg, Emilia Salzmann-Manrique, Andre M. Willasch, and Thomas E. Klingebiel, University Hospital for Children and Adolescents, Frankfurt/Main; Arend von Stackelberg and Cornelia Eckert, Children's Hospital Charité, Berlin, Berlin; Roland Meisel, Heinrich-Heine-University, Düsseldorf; Daniel Stachel, University Hospital Erlangen, Erlangen; Martin Schrappe, Julia Alten, Andre Schrauder, Christian-Albrechts-University and Medical Center Schleswig-Holstein, Kiel; Ansgar Schulz,
| | - Christina Peters
- Peter Bader, Hermann Kreyenberg, Emilia Salzmann-Manrique, Andre M. Willasch, and Thomas E. Klingebiel, University Hospital for Children and Adolescents, Frankfurt/Main; Arend von Stackelberg and Cornelia Eckert, Children's Hospital Charité, Berlin, Berlin; Roland Meisel, Heinrich-Heine-University, Düsseldorf; Daniel Stachel, University Hospital Erlangen, Erlangen; Martin Schrappe, Julia Alten, Andre Schrauder, Christian-Albrechts-University and Medical Center Schleswig-Holstein, Kiel; Ansgar Schulz,
| |
Collapse
|
270
|
Ribera JM, Vives S, Motlló C, Jiménez-Lorenzo MJ. Blinatumomab for treating acute lymphoblastic leukemia. Expert Opin Orphan Drugs 2015. [DOI: 10.1517/21678707.2015.1024223] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
|
271
|
Abstract
Together, acute lymphoblastic leukemia (ALL) and acute myeloid leukemia (AML) make up approximately one-third of all pediatric cancer diagnoses. Despite remarkable improvement in the treatment outcomes of these diseases over the past several decades, the prognosis for certain high-risk groups of leukemia and for relapsed disease remains poor. However, recent insights into different types of 'driver' lesions of leukemogenesis, such as the aberrant activation of signaling pathways and various epigenetic modifications, have led to the discovery of novel agents that specifically target the mechanism of transformation. In parallel, emerging approaches in cancer immunotherapy have led to newer therapies that can exploit and harness cytotoxic immunity directed against malignant cells. This review details the rationale and implementation of recent and specifically targeted therapies in acute pediatric leukemia. Topics covered include the inhibition of critical cell signaling pathways [BCR-ABL, FMS-like tyrosine kinase 3 (FLT3), mammalian target of rapamycin (mTOR), and Janus-associated kinase (JAK)], proteasome inhibition, inhibition of epigenetic regulators of gene expression [DNA methyltransferase (DNMT) inhibitors, histone deacetylase (HDAC) inhibitors, and disruptor of telomeric signaling-1 (DOT1L) inhibitors], monoclonal antibodies and immunoconjugated toxins, bispecific T-cell engaging (BiTE) antibodies, and chimeric antigen receptor-modified (CAR) T cells.
Collapse
Affiliation(s)
- Colleen E Annesley
- Oncology and Pediatrics, The Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Patrick Brown
- Oncology and Pediatrics, The Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, 1650 Orleans Street, CRB-I 2M46, Baltimore, MD 21231, USA
| |
Collapse
|
272
|
Bachireddy P, Burkhardt UE, Rajasagi M, Wu CJ. Haematological malignancies: at the forefront of immunotherapeutic innovation. Nat Rev Cancer 2015; 15:201-15. [PMID: 25786696 PMCID: PMC4511812 DOI: 10.1038/nrc3907] [Citation(s) in RCA: 57] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
The recent successes of cancer immunotherapies have stimulated interest in the potential widespread application of these approaches; haematological malignancies have provided both initial proofs of concept and an informative testing ground for various immune-based therapeutics. The immune-cell origin of many of the blood malignancies provides a unique opportunity both to understand the mechanisms of cancer immune responsiveness and immune evasion, and to exploit these mechanisms for therapeutic purposes.
Collapse
Affiliation(s)
- Pavan Bachireddy
- Department of Medical Oncology and the Cancer Vaccine Center, Dana-Farber Cancer Institute, Boston, MA, USA
- Broad Institute, Cambridge, MA, USA
- Department of Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA, USA
| | - Ute E. Burkhardt
- Department of Medical Oncology and the Cancer Vaccine Center, Dana-Farber Cancer Institute, Boston, MA, USA
| | - Mohini Rajasagi
- Department of Medical Oncology and the Cancer Vaccine Center, Dana-Farber Cancer Institute, Boston, MA, USA
- Broad Institute, Cambridge, MA, USA
| | - Catherine J. Wu
- Department of Medical Oncology and the Cancer Vaccine Center, Dana-Farber Cancer Institute, Boston, MA, USA
- Broad Institute, Cambridge, MA, USA
- Department of Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA, USA
| |
Collapse
|
273
|
Abstract
Despite longstanding efforts in basic research and clinical studies, the prognosis for patients with acute myeloid leukemia (AML) remains poor. About half of the patients are not medically fit for intensive induction therapy to induce a complete remission and are treated with palliative treatment concepts. The patients medically fit for intensive induction therapy have a high complete remission rate but the majority suffers from relapse due to chemo-refractory leukemic cells. Allogeneic stem cell transplantation as post-remission therapy can significantly reduce the likelihood of relapse, but it is associated with a high rate of morbidity and mortality. Novel therapeutic concepts are therefore urgently sought after. During recent years, the focus has shifted towards the development of novel immunotherapeutic strategies. Some of the most promising are drug-conjugated monoclonal antibodies, T-cell engaging antibody constructs, adoptive transfer with chimeric antigen receptor (CAR) T cells, and dendritic cell vaccination. Here, we review recent progress in these four fields and speculate about the optimal time points during the course of AML treatment for their application.
Collapse
Affiliation(s)
- Felix S Lichtenegger
- Department of Internal Medicine III, Klinikum der Universität München, Munich, Germany; Clinical Cooperation Group Immunotherapy at the Helmholtz Institute Munich, Munich, Germany
| | - Christina Krupka
- Department of Internal Medicine III, Klinikum der Universität München, Munich, Germany; Clinical Cooperation Group Immunotherapy at the Helmholtz Institute Munich, Munich, Germany
| | - Thomas Köhnke
- Department of Internal Medicine III, Klinikum der Universität München, Munich, Germany; Clinical Cooperation Group Immunotherapy at the Helmholtz Institute Munich, Munich, Germany
| | - Marion Subklewe
- Department of Internal Medicine III, Klinikum der Universität München, Munich, Germany; Clinical Cooperation Group Immunotherapy at the Helmholtz Institute Munich, Munich, Germany.
| |
Collapse
|
274
|
Lee DW, Kochenderfer JN, Stetler-Stevenson M, Cui YK, Delbrook C, Feldman SA, Fry TJ, Orentas R, Sabatino M, Shah NN, Steinberg SM, Stroncek D, Tschernia N, Yuan C, Zhang H, Zhang L, Rosenberg SA, Wayne AS, Mackall CL. T cells expressing CD19 chimeric antigen receptors for acute lymphoblastic leukaemia in children and young adults: a phase 1 dose-escalation trial. Lancet 2015; 385:517-528. [PMID: 25319501 PMCID: PMC7065359 DOI: 10.1016/s0140-6736(14)61403-3] [Citation(s) in RCA: 2293] [Impact Index Per Article: 229.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
BACKGROUND Chimeric antigen receptor (CAR) modified T cells targeting CD19 have shown activity in case series of patients with acute and chronic lymphocytic leukaemia and B-cell lymphomas, but feasibility, toxicity, and response rates of consecutively enrolled patients treated with a consistent regimen and assessed on an intention-to-treat basis have not been reported. We aimed to define feasibility, toxicity, maximum tolerated dose, response rate, and biological correlates of response in children and young adults with refractory B-cell malignancies treated with CD19-CAR T cells. METHODS This phase 1, dose-escalation trial consecutively enrolled children and young adults (aged 1-30 years) with relapsed or refractory acute lymphoblastic leukaemia or non-Hodgkin lymphoma. Autologous T cells were engineered via an 11-day manufacturing process to express a CD19-CAR incorporating an anti-CD19 single-chain variable fragment plus TCR zeta and CD28 signalling domains. All patients received fludarabine and cyclophosphamide before a single infusion of CD19-CAR T cells. Using a standard 3 + 3 design to establish the maximum tolerated dose, patients received either 1 × 10(6) CAR-transduced T cells per kg (dose 1), 3 × 10(6) CAR-transduced T cells per kg (dose 2), or the entire CAR T-cell product if sufficient numbers of cells to meet the assigned dose were not generated. After the dose-escalation phase, an expansion cohort was treated at the maximum tolerated dose. The trial is registered with ClinicalTrials.gov, number NCT01593696. FINDINGS Between July 2, 2012, and June 20, 2014, 21 patients (including eight who had previously undergone allogeneic haematopoietic stem-cell transplantation) were enrolled and infused with CD19-CAR T cells. 19 received the prescribed dose of CD19-CAR T cells, whereas the assigned dose concentration could not be generated for two patients (90% feasible). All patients enrolled were assessed for response. The maximum tolerated dose was defined as 1 × 10(6) CD19-CAR T cells per kg. All toxicities were fully reversible, with the most severe being grade 4 cytokine release syndrome that occurred in three (14%) of 21 patients (95% CI 3·0-36·3). The most common non-haematological grade 3 adverse events were fever (nine [43%] of 21 patients), hypokalaemia (nine [43%] of 21 patients), fever and neutropenia (eight [38%] of 21 patients), and cytokine release syndrome (three [14%) of 21 patients). INTERPRETATION CD19-CAR T cell therapy is feasible, safe, and mediates potent anti-leukaemic activity in children and young adults with chemotherapy-resistant B-precursor acute lymphoblastic leukaemia. All toxicities were reversible and prolonged B-cell aplasia did not occur. FUNDING National Institutes of Health Intramural funds and St Baldrick's Foundation.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Terry J Fry
- Pediatric Oncology Branch, Bethesda, MD, USA
| | | | - Marianna Sabatino
- Cell Processing Section, Department of Transfusion Medicine, Clinical Center, National Institutes of Health, Bethesda, MD, USA
| | | | - Seth M Steinberg
- Biostatistics and Data Management Section, Office of the Clinical Director, National Cancer Institute, Bethesda, MD, USA
| | - Dave Stroncek
- Cell Processing Section, Department of Transfusion Medicine, Clinical Center, National Institutes of Health, Bethesda, MD, USA
| | - Nick Tschernia
- National Institutes of Health Medical Student Training Program, The University of Nevada School of Medicine, Reno, NV, USA
| | | | - Hua Zhang
- Pediatric Oncology Branch, Bethesda, MD, USA
| | - Ling Zhang
- Pediatric Oncology Branch, Bethesda, MD, USA
| | | | - Alan S Wayne
- Children's Center for Cancer and Blood Diseases, Division of Hematology, Oncology and Blood and Marrow Transplantation, Children's Hospital Los Angeles, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
| | | |
Collapse
|
275
|
Inclusion of an IgG1-Fc spacer abrogates efficacy of CD19 CAR T cells in a xenograft mouse model. Gene Ther 2015; 22:391-403. [DOI: 10.1038/gt.2015.4] [Citation(s) in RCA: 76] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2014] [Revised: 11/03/2014] [Accepted: 01/06/2015] [Indexed: 12/15/2022]
|
276
|
Abstract
The treatment of adult acute lymphoblastic leukemia (ALL) poses a tremendous challenge for hematologists. The use of pediatric-based chemotherapy regimens in young adults up to the age of 45 years has resulted in improved outcomes when compared retrospectively with historical controls treated with adult therapy. A better understanding of the molecular landscape of ALL and advances in the field of monoclonal antibody therapy have resulted in the development of several new agents that may provide for a reduction in the toxicity inherent in pediatric-like regimens. The anti-CD20 antibody, rituximab, anti CD22 antibody, epratuzumab, anti-CD22 antibody-drug conjugate, Inotuzumab ozogamicin, the bi-specific T-cell engager (BiTE) antibody, Blinatumomab, and chimeric receptor antigen (CAR) therapy are among the emerging agents that have demonstrated the potential to improve response rate and decrease toxicity when used alone or in combination with chemotherapy. Several role-defining phase II and phase III clinical trials with these agents are currently underway in the relapsed/refractory and newly diagnosed ALL settings.
Collapse
|
277
|
Vacchelli E, Pol J, Bloy N, Eggermont A, Cremer I, Fridman WH, Galon J, Marabelle A, Kohrt H, Zitvogel L, Kroemer G, Galluzzi L. Trial watch: Tumor-targeting monoclonal antibodies for oncological indications. Oncoimmunology 2015; 4:e985940. [PMID: 25949870 DOI: 10.4161/2162402x.2014.985940] [Citation(s) in RCA: 42] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2014] [Accepted: 05/11/2014] [Indexed: 12/31/2022] Open
Abstract
An expanding panel of monoclonal antibodies (mAbs) that specifically target malignant cells or intercept trophic factors delivered by the tumor stroma is now available for cancer therapy. These mAbs can exert direct antiproliferative/cytotoxic effects as they inhibit pro-survival signal transduction cascades or activate lethal receptors at the plasma membrane of cancer cells, they can opsonize neoplastic cells to initiate a tumor-targeting immune response, or they can be harnessed to specifically deliver toxins or radionuclides to transformed cells. As an indication of the success of this immunotherapeutic paradigm, international regulatory agencies approve new tumor-targeting mAbs for use in cancer patients every year. Moreover, the list of indications for previously licensed molecules is frequently expanded to other neoplastic disorders as the results of large, randomized clinical trials become available. Here, we discuss recent advances in the preclinical and clinical development of tumor-targeting mAbs for oncological indications.
Collapse
Affiliation(s)
- Erika Vacchelli
- Gustave Roussy Cancer Campus ; Villejuif, France ; INSERM; U1138 ; Paris, France ; Equipe 11 labellisée par la Ligue Nationale contre le Cancer; Centre de Recherche des Cordeliers ; Paris, France
| | - Jonathan Pol
- Gustave Roussy Cancer Campus ; Villejuif, France ; INSERM; U1138 ; Paris, France ; Equipe 11 labellisée par la Ligue Nationale contre le Cancer; Centre de Recherche des Cordeliers ; Paris, France
| | - Norma Bloy
- Gustave Roussy Cancer Campus ; Villejuif, France ; INSERM; U1138 ; Paris, France ; Equipe 11 labellisée par la Ligue Nationale contre le Cancer; Centre de Recherche des Cordeliers ; Paris, France
| | | | - Isabelle Cremer
- INSERM; U1138 ; Paris, France ; Equipe 13; Centre de Recherche des Cordeliers ; Paris, France ; Université Pierre et Marie Curie/Paris VI ; Paris, France
| | - Wolf Hervé Fridman
- INSERM; U1138 ; Paris, France ; Equipe 13; Centre de Recherche des Cordeliers ; Paris, France ; Université Pierre et Marie Curie/Paris VI ; Paris, France
| | - Jérôme Galon
- INSERM; U1138 ; Paris, France ; Université Pierre et Marie Curie/Paris VI ; Paris, France ; Laboratory of Integrative Cancer Immunology; Centre de Recherche des Cordeliers ; Paris, France ; Université Paris Descartes/Paris V; Sorbonne Paris Cité ; Paris, France
| | - Aurélien Marabelle
- Gustave Roussy Cancer Campus ; Villejuif, France ; INSERM ; U1015 , Villejuif, France
| | - Holbrook Kohrt
- Department of Medicine; Division of Oncology; Stanford University ; Stanford, CA, USA
| | - Laurence Zitvogel
- Gustave Roussy Cancer Campus ; Villejuif, France ; INSERM ; U1015 , Villejuif, France
| | - Guido Kroemer
- INSERM; U1138 ; Paris, France ; Equipe 11 labellisée par la Ligue Nationale contre le Cancer; Centre de Recherche des Cordeliers ; Paris, France ; Université Paris Descartes/Paris V; Sorbonne Paris Cité ; Paris, France ; Pôle de Biologie; Hôpital Européen Georges Pompidou ; AP-HP ; Paris, France ; Metabolomics and Cell Biology Platforms; Gustave Roussy Cancer Campus ; Villejuif, France
| | - Lorenzo Galluzzi
- Gustave Roussy Cancer Campus ; Villejuif, France ; INSERM; U1138 ; Paris, France ; Equipe 11 labellisée par la Ligue Nationale contre le Cancer; Centre de Recherche des Cordeliers ; Paris, France ; Université Paris Descartes/Paris V; Sorbonne Paris Cité ; Paris, France
| |
Collapse
|
278
|
Maino E, Scattolin AM, Viero P, Sancetta R, Pascarella A, Vespignani M, Bassan R. Modern immunotherapy of adult B-lineage acute lymphoblastic leukemia with monoclonal antibodies and chimeric antigen receptor modified T cells. Mediterr J Hematol Infect Dis 2015; 7:e2015001. [PMID: 25574360 PMCID: PMC4283921 DOI: 10.4084/mjhid.2015.001] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2014] [Accepted: 11/20/2014] [Indexed: 11/08/2022] Open
Abstract
The introduction of newer cytotoxic monoclonal antibodies and chimeric antigen receptor modified T cells is opening a new age in the management of B-lineage adult acute lymphoblastic leukemia. This therapeutic change must be very positively acknowledged because of the limits of intensive chemotherapy programs and allogeneic stem cell transplantation. In fact, with these traditional therapeutic tools the cure can be achieved in only 40-50% of the patients. The failure rates are particularly high in the elderly, in patients with post-induction persistence of minimal residual disease and especially in refractory/relapsed disease. The place of the novel immunotherapeutics in improving the outcome of adult patients with B-lineage acute lymphoblastic leukemia is reviewed.
Collapse
Affiliation(s)
- Elena Maino
- Hematology and Bone Marrow Transplant Unit, Ospedale dell’Angelo e SS. Giovanni e Paolo, Mestre-Venezia, Italy
| | - Anna Maria Scattolin
- Hematology and Bone Marrow Transplant Unit, Ospedale dell’Angelo e SS. Giovanni e Paolo, Mestre-Venezia, Italy
| | - Piera Viero
- Hematology and Bone Marrow Transplant Unit, Ospedale dell’Angelo e SS. Giovanni e Paolo, Mestre-Venezia, Italy
| | - Rosaria Sancetta
- Hematology and Bone Marrow Transplant Unit, Ospedale dell’Angelo e SS. Giovanni e Paolo, Mestre-Venezia, Italy
| | - Anna Pascarella
- Hematology and Bone Marrow Transplant Unit, Ospedale dell’Angelo e SS. Giovanni e Paolo, Mestre-Venezia, Italy
| | - Michele Vespignani
- Hematology and Bone Marrow Transplant Unit, Ospedale dell’Angelo e SS. Giovanni e Paolo, Mestre-Venezia, Italy
| | - Renato Bassan
- Hematology and Bone Marrow Transplant Unit, Ospedale dell’Angelo e SS. Giovanni e Paolo, Mestre-Venezia, Italy
| |
Collapse
|
279
|
Prognostic Factors in Adolescent and Adult Patients With Acute Lymphoblastic Leukemia With Two Protocols of Chemotherapy: A Cross-Sectional Study. CLINICAL LYMPHOMA MYELOMA & LEUKEMIA 2015; 15:e7-e14. [DOI: 10.1016/j.clml.2014.07.006] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/15/2014] [Revised: 07/03/2014] [Accepted: 07/08/2014] [Indexed: 11/18/2022]
|
280
|
Shalabi H, Angiolillo A, Fry TJ. Beyond CD19: Opportunities for Future Development of Targeted Immunotherapy in Pediatric Relapsed-Refractory Acute Leukemia. Front Pediatr 2015; 3:80. [PMID: 26484338 PMCID: PMC4589648 DOI: 10.3389/fped.2015.00080] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/17/2015] [Accepted: 09/18/2015] [Indexed: 12/30/2022] Open
Abstract
Chimeric antigen receptor (CAR) T cell therapy has been used as a targeted approach in cancer therapy. Relapsed and refractory acute leukemia in pediatrics has been difficult to treat with conventional therapy due to dose-limiting toxicities. With the recent success of CD 19 CAR in pediatric patients with B cell acute lymphoblastic leukemia (ALL), this mode of therapy has become a very attractive option for these patients with high-risk disease. In this review, we will discuss current treatment paradigms of pediatric acute leukemia and potential therapeutic targets for additional high-risk populations, including T cell ALL, AML, and infant ALL.
Collapse
Affiliation(s)
- Haneen Shalabi
- Center for Cancer and Blood Disorders, Children's National Medical Center , Washington, DC , USA
| | - Anne Angiolillo
- Center for Cancer and Blood Disorders, Children's National Medical Center , Washington, DC , USA
| | - Terry J Fry
- Hematologic Malignancies Section, Pediatric Oncology Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health , Bethesda, MD , USA
| |
Collapse
|
281
|
Galluzzi L, Vacchelli E, Pedro JMBS, Buqué A, Senovilla L, Baracco EE, Bloy N, Castoldi F, Abastado JP, Agostinis P, Apte RN, Aranda F, Ayyoub M, Beckhove P, Blay JY, Bracci L, Caignard A, Castelli C, Cavallo F, Celis E, Cerundolo V, Clayton A, Colombo MP, Coussens L, Dhodapkar MV, Eggermont AM, Fearon DT, Fridman WH, Fučíková J, Gabrilovich DI, Galon J, Garg A, Ghiringhelli F, Giaccone G, Gilboa E, Gnjatic S, Hoos A, Hosmalin A, Jäger D, Kalinski P, Kärre K, Kepp O, Kiessling R, Kirkwood JM, Klein E, Knuth A, Lewis CE, Liblau R, Lotze MT, Lugli E, Mach JP, Mattei F, Mavilio D, Melero I, Melief CJ, Mittendorf EA, Moretta L, Odunsi A, Okada H, Palucka AK, Peter ME, Pienta KJ, Porgador A, Prendergast GC, Rabinovich GA, Restifo NP, Rizvi N, Sautès-Fridman C, Schreiber H, Seliger B, Shiku H, Silva-Santos B, Smyth MJ, Speiser DE, Spisek R, Srivastava PK, Talmadge JE, Tartour E, Van Der Burg SH, Van Den Eynde BJ, Vile R, Wagner H, Weber JS, Whiteside TL, Wolchok JD, Zitvogel L, Zou W, Kroemer G. Classification of current anticancer immunotherapies. Oncotarget 2014; 5:12472-508. [PMID: 25537519 PMCID: PMC4350348 DOI: 10.18632/oncotarget.2998] [Citation(s) in RCA: 339] [Impact Index Per Article: 30.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2014] [Accepted: 12/15/2014] [Indexed: 11/25/2022] Open
Abstract
During the past decades, anticancer immunotherapy has evolved from a promising therapeutic option to a robust clinical reality. Many immunotherapeutic regimens are now approved by the US Food and Drug Administration and the European Medicines Agency for use in cancer patients, and many others are being investigated as standalone therapeutic interventions or combined with conventional treatments in clinical studies. Immunotherapies may be subdivided into "passive" and "active" based on their ability to engage the host immune system against cancer. Since the anticancer activity of most passive immunotherapeutics (including tumor-targeting monoclonal antibodies) also relies on the host immune system, this classification does not properly reflect the complexity of the drug-host-tumor interaction. Alternatively, anticancer immunotherapeutics can be classified according to their antigen specificity. While some immunotherapies specifically target one (or a few) defined tumor-associated antigen(s), others operate in a relatively non-specific manner and boost natural or therapy-elicited anticancer immune responses of unknown and often broad specificity. Here, we propose a critical, integrated classification of anticancer immunotherapies and discuss the clinical relevance of these approaches.
Collapse
Affiliation(s)
- Lorenzo Galluzzi
- Equipe 11 labellisée pas la Ligue Nationale contre le Cancer, Centre de Recherche des Cordeliers, Paris, France
- INSERM, U1138, Paris, France
- Gustave Roussy Cancer Campus, Villejuif, France
- Université Paris Descartes/Paris V, Sorbonne Paris Cité, Paris, France
| | - Erika Vacchelli
- Equipe 11 labellisée pas la Ligue Nationale contre le Cancer, Centre de Recherche des Cordeliers, Paris, France
- INSERM, U1138, Paris, France
- Gustave Roussy Cancer Campus, Villejuif, France
| | - José-Manuel Bravo-San Pedro
- Equipe 11 labellisée pas la Ligue Nationale contre le Cancer, Centre de Recherche des Cordeliers, Paris, France
- INSERM, U1138, Paris, France
- Gustave Roussy Cancer Campus, Villejuif, France
| | - Aitziber Buqué
- Equipe 11 labellisée pas la Ligue Nationale contre le Cancer, Centre de Recherche des Cordeliers, Paris, France
- INSERM, U1138, Paris, France
- Gustave Roussy Cancer Campus, Villejuif, France
| | - Laura Senovilla
- Equipe 11 labellisée pas la Ligue Nationale contre le Cancer, Centre de Recherche des Cordeliers, Paris, France
- INSERM, U1138, Paris, France
- Gustave Roussy Cancer Campus, Villejuif, France
| | - Elisa Elena Baracco
- Equipe 11 labellisée pas la Ligue Nationale contre le Cancer, Centre de Recherche des Cordeliers, Paris, France
- INSERM, U1138, Paris, France
- Gustave Roussy Cancer Campus, Villejuif, France
- Faculté de Medicine, Université Paris Sud/Paris XI, Le Kremlin-Bicêtre, France
| | - Norma Bloy
- Equipe 11 labellisée pas la Ligue Nationale contre le Cancer, Centre de Recherche des Cordeliers, Paris, France
- INSERM, U1138, Paris, France
- Gustave Roussy Cancer Campus, Villejuif, France
- Faculté de Medicine, Université Paris Sud/Paris XI, Le Kremlin-Bicêtre, France
| | - Francesca Castoldi
- Equipe 11 labellisée pas la Ligue Nationale contre le Cancer, Centre de Recherche des Cordeliers, Paris, France
- INSERM, U1138, Paris, France
- Gustave Roussy Cancer Campus, Villejuif, France
- Faculté de Medicine, Université Paris Sud/Paris XI, Le Kremlin-Bicêtre, France
- Sotio a.c., Prague, Czech Republic
| | - Jean-Pierre Abastado
- Pole d'innovation thérapeutique en oncologie, Institut de Recherches Internationales Servier, Suresnes, France
| | - Patrizia Agostinis
- Cell Death Research and Therapy (CDRT) Laboratory, Dept. of Cellular and Molecular Medicine, University of Leuven, Leuven, Belgium
| | - Ron N. Apte
- The Shraga Segal Dept. of Microbiology, Immunology and Genetics, Faculty of Health Sciences, Ben-Gurion University of the Negev, Beer-Sheva, Israel
| | - Fernando Aranda
- Equipe 11 labellisée pas la Ligue Nationale contre le Cancer, Centre de Recherche des Cordeliers, Paris, France
- INSERM, U1138, Paris, France
- Gustave Roussy Cancer Campus, Villejuif, France
- Group of Immune receptors of the Innate and Adaptive System, Institut d'Investigacions Biomédiques August Pi i Sunyer (IDIBAPS), Barcelona, Spain
| | - Maha Ayyoub
- INSERM, U1102, Saint Herblain, France
- Institut de Cancérologie de l'Ouest, Saint Herblain, France
| | - Philipp Beckhove
- Translational Immunology Division, German Cancer Research Center, Heidelberg, Germany
| | - Jean-Yves Blay
- Equipe 11, Centre Léon Bérard (CLR), Lyon, France
- Centre de Recherche en Cancérologie de Lyon (CRCL), Lyon, France
| | - Laura Bracci
- Dept. of Hematology, Oncology and Molecular Medicine, Istituto Superiore di Sanità, Rome, Italy
| | - Anne Caignard
- INSERM, U1160, Paris, France
- Groupe Hospitalier Saint Louis-Lariboisière - F. Vidal, Paris, France
| | - Chiara Castelli
- Unit of Immunotherapy of Human Tumors, Dept. of Experimental Oncology and Molecular Medicine, Fondazione IRCCS Istituto Nazionale Tumori, Milano, Italy
| | - Federica Cavallo
- Molecular Biotechnology Center, Dept. of Molecular Biotechnology and Health Sciences, University of Torino, Torino, Italy
| | - Estaban Celis
- Cancer Immunology, Inflammation and Tolerance Program, Georgia Regents University Cancer Center, Augusta, GA, USA
| | - Vincenzo Cerundolo
- MRC Human Immunology Unit, Weatherall Institute of Molecular Medicine, University of Oxford, Oxford, UK
| | - Aled Clayton
- Institute of Cancer & Genetics, School of Medicine, Cardiff University, Cardiff, UK
- Velindre Cancer Centre, Cardiff, UK
| | - Mario P. Colombo
- Unit of Immunotherapy of Human Tumors, Dept. of Experimental Oncology and Molecular Medicine, Fondazione IRCCS Istituto Nazionale Tumori, Milano, Italy
| | - Lisa Coussens
- Knight Cancer Institute, Oregon Health & Science University, Portland, OR, USA
| | - Madhav V. Dhodapkar
- Sect. of Hematology and Immunobiology, Yale Cancer Center, Yale University, New Haven, CT, USA
| | | | | | - Wolf H. Fridman
- INSERM, U1138, Paris, France
- Université Paris Descartes/Paris V, Sorbonne Paris Cité, Paris, France
- Université Pierre et Marie Curie/Paris VI, Paris, France
- Equipe 13, Centre de Recherche des Cordeliers, Paris, France
| | - Jitka Fučíková
- Sotio a.c., Prague, Czech Republic
- Dept. of Immunology, 2nd Faculty of Medicine and University Hospital Motol, Charles University, Prague, Czech Republic
| | - Dmitry I. Gabrilovich
- Dept. of Pathology and Laboratory Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Jérôme Galon
- INSERM, U1138, Paris, France
- Université Paris Descartes/Paris V, Sorbonne Paris Cité, Paris, France
- Université Pierre et Marie Curie/Paris VI, Paris, France
- Laboratory of Integrative Cancer Immunology, Centre de Recherche des Cordeliers, Paris, France
| | - Abhishek Garg
- Cell Death Research and Therapy (CDRT) Laboratory, Dept. of Cellular and Molecular Medicine, University of Leuven, Leuven, Belgium
| | - François Ghiringhelli
- INSERM, UMR866, Dijon, France
- Centre Georges François Leclerc, Dijon, France
- Université de Bourgogne, Dijon, France
| | - Giuseppe Giaccone
- Center for Cancer Research, National Cancer Institute (NCI), National Institutes of Health (NIH), Bethesda, MD, USA
- Lombardi Comprehensive Cancer Center, Georgetown University, Washington, DC, USA
| | - Eli Gilboa
- Dept. of Microbiology and Immunology, Sylvester Comprehensive Cancer Center, University of Miami, Miller School of Medicine, Miami, FL, USA
| | - Sacha Gnjatic
- Sect. of Hematology/Oncology, Immunology, Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Axel Hoos
- Glaxo Smith Kline, Cancer Immunotherapy Consortium, Collegeville, PA, USA
| | - Anne Hosmalin
- Université Paris Descartes/Paris V, Sorbonne Paris Cité, Paris, France
- INSERM, U1016, Paris, France
- CNRS, UMR8104, Paris, France
- Hôpital Cochin, AP-HP, Paris, France
| | - Dirk Jäger
- National Center for Tumor Diseases, University Medical Center Heidelberg, Heidelberg, Germany
| | - Pawel Kalinski
- Dept. of Surgery, University of Pittsburgh, Pittsburgh, PA, USA
- University of Pittsburgh Cancer Institute, Hillman Cancer Center, Pittsburgh, PA, USA
- Dept. of Immunology and Infectious Diseases and Microbiology, University of Pittsburgh, Pittsburgh, PA, USA
| | - Klas Kärre
- Dept. of Microbiology, Tumor and Cell Biology, Karolinska Institute, Stockholm, Sweden
| | - Oliver Kepp
- Equipe 11 labellisée pas la Ligue Nationale contre le Cancer, Centre de Recherche des Cordeliers, Paris, France
- INSERM, U1138, Paris, France
- Metabolomics and Cell Biology Platforms, Gustave Roussy Cancer Campus, Villejuif, France
| | - Rolf Kiessling
- Dept. of Oncology, Karolinska Institute Hospital, Stockholm, Sweden
| | - John M. Kirkwood
- University of Pittsburgh Cancer Institute Laboratory, Pittsburgh, PA, USA
| | - Eva Klein
- Dept. of Microbiology, Tumor and Cell Biology, Karolinska Institute, Stockholm, Sweden
| | - Alexander Knuth
- National Center for Cancer Care and Research, Hamad Medical Corporation, Doha, Qatar
| | - Claire E. Lewis
- Academic Unit of Inflammation and Tumour Targeting, Dept. of Oncology, University of Sheffield Medical School, Sheffield, UK
| | - Roland Liblau
- INSERM, UMR1043, Toulouse, France
- CNRS, UMR5282, Toulouse, France
- Laboratoire d'Immunologie, CHU Toulouse, Université Toulouse II, Toulouse, France
| | - Michael T. Lotze
- Dept. of Surgery, University of Pittsburgh, Pittsburgh, PA, USA
- University of Pittsburgh Cancer Institute, Hillman Cancer Center, Pittsburgh, PA, USA
| | - Enrico Lugli
- Unit of Clinical and Experimental Immunology, Humanitas Clinical and Research Institute, Rozzano, Italy
| | - Jean-Pierre Mach
- Dept. of Biochemistry, University of Lausanne, Epalinges, Switzerland
| | - Fabrizio Mattei
- Dept. of Hematology, Oncology and Molecular Medicine, Istituto Superiore di Sanità, Rome, Italy
| | - Domenico Mavilio
- Unit of Clinical and Experimental Immunology, Humanitas Clinical and Research Institute, Rozzano, Italy
- Dept. of Medical Biotechnologies and Translational Medicine, University of Milan, Rozzano, Italy
| | - Ignacio Melero
- Dept. of Immunology, Centro de Investigación Médica Aplicada (CIMA), Universidad de Navarra, Pamplona, Spain
- Dept. of Oncology, Clínica Universidad de Navarra, Pamplona, Spain
| | - Cornelis J. Melief
- ISA Therapeutics, Leiden, The Netherlands
- Dept. of Immunohematology and Blood Transfusion, Leiden University Medical Center, Leiden, The Netherlands
| | - Elizabeth A. Mittendorf
- Research Dept. of Surgical Oncology, The University of Texas, MD Anderson Cancer Center, Houston, TX, USA
| | | | - Adekunke Odunsi
- Center for Immunotherapy, Roswell Park Cancer Institute, Buffalo, NY, USA
| | - Hideho Okada
- Dept. of Neurological Surgery, University of California San Francisco, San Francisco, CA, USA
| | | | - Marcus E. Peter
- Div. of Hematology/Oncology, Northwestern University, Feinberg School of Medicine, Chicago, IL, USA
| | - Kenneth J. Pienta
- The James Buchanan Brady Urological Institute, The Johns Hopkins Medical Institutions, Baltimore, MD, USA
| | - Angel Porgador
- The Shraga Segal Dept. of Microbiology, Immunology and Genetics, Faculty of Health Sciences, Ben-Gurion University of the Negev, Beer-Sheva, Israel
| | - George C. Prendergast
- Lankenau Institute for Medical Research, Wynnewood, PA, USA
- Dept. of Pathology, Anatomy and Cell Biology, Sidney Kimmel Medical College, Philadelphia, PA, USA
- Cell Biology and Signaling Program, Kimmel Cancer Center, Thomas Jefferson University, Philadelphia, PA, USA
| | - Gabriel A. Rabinovich
- Laboratorio de Inmunopatología, Instituto de Biología y Medicina Experimental (IBYME), Buenos Aires, Argentina
| | - Nicholas P. Restifo
- National Cancer Institute (NCI), National Institutes of Health (NIH), Bethesda, MD, USA
| | - Naiyer Rizvi
- Memorial Sloan Kettering Cancer Center (MSKCC), New York, NY, USA
| | - Catherine Sautès-Fridman
- INSERM, U1138, Paris, France
- Université Paris Descartes/Paris V, Sorbonne Paris Cité, Paris, France
- Université Pierre et Marie Curie/Paris VI, Paris, France
- Equipe 13, Centre de Recherche des Cordeliers, Paris, France
| | - Hans Schreiber
- Dept. of Pathology, The Cancer Research Center, The University of Chicago, Chicago, IL, USA
| | - Barbara Seliger
- Institute of Medical Immunology, Martin Luther University Halle-Wittenberg, Halle, Germany
| | - Hiroshi Shiku
- Dept. of Immuno-GeneTherapy, Mie University Graduate School of Medicine, Tsu, Japan
| | - Bruno Silva-Santos
- Instituto de Medicina Molecular, Universidade de Lisboa, Lisboa, Portugal
| | - Mark J. Smyth
- Immunology in Cancer and Infection Laboratory, QIMR Berghofer Medical Research Institute, Herston, Queensland, Australia
- School of Medicine, University of Queensland, Herston, Queensland, Australia
| | - Daniel E. Speiser
- Dept. of Oncology, University of Lausanne, Lausanne, Switzerland
- Ludwig Cancer Research Center, Lausanne, Switzerland
| | - Radek Spisek
- Sotio a.c., Prague, Czech Republic
- Dept. of Immunology, 2nd Faculty of Medicine and University Hospital Motol, Charles University, Prague, Czech Republic
| | - Pramod K. Srivastava
- Dept. of Immunology, University of Connecticut School of Medicine, Farmington, CT, USA
- Carole and Ray Neag Comprehensive Cancer Center, Farmington, CT, USA
| | - James E. Talmadge
- Laboratory of Transplantation Immunology, Dept. of Pathology and Microbiology, University of Nebraska Medical Center, Omaha, NE, USA
| | - Eric Tartour
- Université Paris Descartes/Paris V, Sorbonne Paris Cité, Paris, France
- INSERM, U970, Paris, France
- Paris-Cardiovascular Research Center (PARCC), Paris, France
- Service d'Immunologie Biologique, Hôpital Européen Georges Pompidou (HEGP), AP-HP, Paris, France
| | | | - Benoît J. Van Den Eynde
- Ludwig Institute for Cancer Research, Brussels, Belgium
- de Duve Institute, Brussels, Belgium
- Université Catholique de Louvain, Brussels, Belgium
| | - Richard Vile
- Dept. of Molecular Medicine and Immunology, Mayo Clinic College of Medicine, Rochester, MN, USA
| | - Hermann Wagner
- Institute of Medical Microbiology, Immunology and Hygiene, Technical University Munich, Munich, Germany
| | - Jeffrey S. Weber
- Donald A. Adam Comprehensive Melanoma Research Center, Moffitt Cancer Center, Tampa, FL, USA
| | - Theresa L. Whiteside
- University of Pittsburgh Cancer Institute, Hillman Cancer Center, Pittsburgh, PA, USA
- University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Jedd D. Wolchok
- Dept. of Medicine and Ludwig Center, Memorial Sloan Kettering Cancer Center (MSKCC), New York, NY, USA
- Weill Cornell Medical College, New York, NY, USA
| | - Laurence Zitvogel
- Gustave Roussy Cancer Campus, Villejuif, France
- INSERM, U1015, Villejuif, France
- Centre d'Investigation Clinique Biothérapie 507 (CICBT507), Gustave Roussy Cancer Campus, Villejuif, France
| | - Weiping Zou
- University of Michigan, School of Medicine, Ann Arbor, MI, USA
| | - Guido Kroemer
- Equipe 11 labellisée pas la Ligue Nationale contre le Cancer, Centre de Recherche des Cordeliers, Paris, France
- INSERM, U1138, Paris, France
- Université Paris Descartes/Paris V, Sorbonne Paris Cité, Paris, France
- Metabolomics and Cell Biology Platforms, Gustave Roussy Cancer Campus, Villejuif, France
- Pôle de Biologie, Hôpital Européen Georges Pompidou (HEGP), AP-HP, Paris, France
| |
Collapse
|
282
|
Topp MS, Gökbuget N, Zugmaier G, Klappers P, Stelljes M, Neumann S, Viardot A, Marks R, Diedrich H, Faul C, Reichle A, Horst HA, Brüggemann M, Wessiepe D, Holland C, Alekar S, Mergen N, Einsele H, Hoelzer D, Bargou RC. Phase II Trial of the Anti-CD19 Bispecific T Cell–Engager Blinatumomab Shows Hematologic and Molecular Remissions in Patients With Relapsed or Refractory B-Precursor Acute Lymphoblastic Leukemia. J Clin Oncol 2014; 32:4134-40. [DOI: 10.1200/jco.2014.56.3247] [Citation(s) in RCA: 482] [Impact Index Per Article: 43.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Purpose Patients with relapsed or refractory acute lymphoblastic leukemia (ALL) have a dismal prognosis. CD19 is homogenously expressed in B-precursor ALL and can be targeted by the investigational bispecific T cell–engager antibody blinatumomab. A phase II trial was performed to determine clinical activity in this patient cohort. Patients and Methods Thirty-six patients with relapsed or refractory B-precursor ALL were treated with blinatumomab in cycles of 4-week continuous infusion followed by a 2-week treatment-free interval in a single-arm study with a dose-finding stage and an extension stage. The primary end point was complete remission (CR) or CR with partial hematologic recovery (CRh). Major secondary end points included minimal residual disease (MRD) response, rate of allogeneic hematopoietic stem-cell transplantation (HSCT) realization, relapse-free survival (RFS), overall survival (OS), and incidence of adverse events (AEs). Results Median age was 32 years (range, 18 to 77 years). Twenty-five patients (69%) achieved a CR or CRh, with 88% of the responders achieving an MRD response. Median OS was 9.8 months (95% CI, 8.5 to 14.9), and median RFS was 7.6 months (95% CI, 4.5 to 9.5). Thirteen responders (52%) underwent HSCT after achieving a CR or CRh. The most frequent AE during treatment was pyrexia (grade 1 or 2, 75%; grade 3, 6%). In six patients with nervous system or psychiatric disorder AEs and in two patients with cytokine release syndrome, treatment had to be interrupted or discontinued. These medical events were resolved clinically. Conclusion The data support further investigation of blinatumomab for the treatment of adult patients with relapsed or refractory ALL in a larger confirmatory study.
Collapse
Affiliation(s)
- Max S. Topp
- Max S. Topp, Hermann Einsele, and Ralf C. Bargou, Universitätsklinikum Würzburg, Würzburg; Nicola Gökbuget, Goethe University; Dieter Hoelzer, Onkologikum, Frankfurt; Gerhard Zugmaier, Petra Klappers, and Noemi Mergen, Amgen Research; Dorothea Wessiepe, Metronomia, Munich; Matthias Stelljes, University of Münster, Münster; Svenja Neumann, Heinz-August Horst, and Monika Brüggemann, University Schleswig Holstein, City Hospital, Kiel; Andreas Viardot, University of Ulm, Ulm; Reinhard Marks,
| | - Nicola Gökbuget
- Max S. Topp, Hermann Einsele, and Ralf C. Bargou, Universitätsklinikum Würzburg, Würzburg; Nicola Gökbuget, Goethe University; Dieter Hoelzer, Onkologikum, Frankfurt; Gerhard Zugmaier, Petra Klappers, and Noemi Mergen, Amgen Research; Dorothea Wessiepe, Metronomia, Munich; Matthias Stelljes, University of Münster, Münster; Svenja Neumann, Heinz-August Horst, and Monika Brüggemann, University Schleswig Holstein, City Hospital, Kiel; Andreas Viardot, University of Ulm, Ulm; Reinhard Marks,
| | - Gerhard Zugmaier
- Max S. Topp, Hermann Einsele, and Ralf C. Bargou, Universitätsklinikum Würzburg, Würzburg; Nicola Gökbuget, Goethe University; Dieter Hoelzer, Onkologikum, Frankfurt; Gerhard Zugmaier, Petra Klappers, and Noemi Mergen, Amgen Research; Dorothea Wessiepe, Metronomia, Munich; Matthias Stelljes, University of Münster, Münster; Svenja Neumann, Heinz-August Horst, and Monika Brüggemann, University Schleswig Holstein, City Hospital, Kiel; Andreas Viardot, University of Ulm, Ulm; Reinhard Marks,
| | - Petra Klappers
- Max S. Topp, Hermann Einsele, and Ralf C. Bargou, Universitätsklinikum Würzburg, Würzburg; Nicola Gökbuget, Goethe University; Dieter Hoelzer, Onkologikum, Frankfurt; Gerhard Zugmaier, Petra Klappers, and Noemi Mergen, Amgen Research; Dorothea Wessiepe, Metronomia, Munich; Matthias Stelljes, University of Münster, Münster; Svenja Neumann, Heinz-August Horst, and Monika Brüggemann, University Schleswig Holstein, City Hospital, Kiel; Andreas Viardot, University of Ulm, Ulm; Reinhard Marks,
| | - Matthias Stelljes
- Max S. Topp, Hermann Einsele, and Ralf C. Bargou, Universitätsklinikum Würzburg, Würzburg; Nicola Gökbuget, Goethe University; Dieter Hoelzer, Onkologikum, Frankfurt; Gerhard Zugmaier, Petra Klappers, and Noemi Mergen, Amgen Research; Dorothea Wessiepe, Metronomia, Munich; Matthias Stelljes, University of Münster, Münster; Svenja Neumann, Heinz-August Horst, and Monika Brüggemann, University Schleswig Holstein, City Hospital, Kiel; Andreas Viardot, University of Ulm, Ulm; Reinhard Marks,
| | - Svenja Neumann
- Max S. Topp, Hermann Einsele, and Ralf C. Bargou, Universitätsklinikum Würzburg, Würzburg; Nicola Gökbuget, Goethe University; Dieter Hoelzer, Onkologikum, Frankfurt; Gerhard Zugmaier, Petra Klappers, and Noemi Mergen, Amgen Research; Dorothea Wessiepe, Metronomia, Munich; Matthias Stelljes, University of Münster, Münster; Svenja Neumann, Heinz-August Horst, and Monika Brüggemann, University Schleswig Holstein, City Hospital, Kiel; Andreas Viardot, University of Ulm, Ulm; Reinhard Marks,
| | - Andreas Viardot
- Max S. Topp, Hermann Einsele, and Ralf C. Bargou, Universitätsklinikum Würzburg, Würzburg; Nicola Gökbuget, Goethe University; Dieter Hoelzer, Onkologikum, Frankfurt; Gerhard Zugmaier, Petra Klappers, and Noemi Mergen, Amgen Research; Dorothea Wessiepe, Metronomia, Munich; Matthias Stelljes, University of Münster, Münster; Svenja Neumann, Heinz-August Horst, and Monika Brüggemann, University Schleswig Holstein, City Hospital, Kiel; Andreas Viardot, University of Ulm, Ulm; Reinhard Marks,
| | - Reinhard Marks
- Max S. Topp, Hermann Einsele, and Ralf C. Bargou, Universitätsklinikum Würzburg, Würzburg; Nicola Gökbuget, Goethe University; Dieter Hoelzer, Onkologikum, Frankfurt; Gerhard Zugmaier, Petra Klappers, and Noemi Mergen, Amgen Research; Dorothea Wessiepe, Metronomia, Munich; Matthias Stelljes, University of Münster, Münster; Svenja Neumann, Heinz-August Horst, and Monika Brüggemann, University Schleswig Holstein, City Hospital, Kiel; Andreas Viardot, University of Ulm, Ulm; Reinhard Marks,
| | - Helmut Diedrich
- Max S. Topp, Hermann Einsele, and Ralf C. Bargou, Universitätsklinikum Würzburg, Würzburg; Nicola Gökbuget, Goethe University; Dieter Hoelzer, Onkologikum, Frankfurt; Gerhard Zugmaier, Petra Klappers, and Noemi Mergen, Amgen Research; Dorothea Wessiepe, Metronomia, Munich; Matthias Stelljes, University of Münster, Münster; Svenja Neumann, Heinz-August Horst, and Monika Brüggemann, University Schleswig Holstein, City Hospital, Kiel; Andreas Viardot, University of Ulm, Ulm; Reinhard Marks,
| | - Christoph Faul
- Max S. Topp, Hermann Einsele, and Ralf C. Bargou, Universitätsklinikum Würzburg, Würzburg; Nicola Gökbuget, Goethe University; Dieter Hoelzer, Onkologikum, Frankfurt; Gerhard Zugmaier, Petra Klappers, and Noemi Mergen, Amgen Research; Dorothea Wessiepe, Metronomia, Munich; Matthias Stelljes, University of Münster, Münster; Svenja Neumann, Heinz-August Horst, and Monika Brüggemann, University Schleswig Holstein, City Hospital, Kiel; Andreas Viardot, University of Ulm, Ulm; Reinhard Marks,
| | - Albrecht Reichle
- Max S. Topp, Hermann Einsele, and Ralf C. Bargou, Universitätsklinikum Würzburg, Würzburg; Nicola Gökbuget, Goethe University; Dieter Hoelzer, Onkologikum, Frankfurt; Gerhard Zugmaier, Petra Klappers, and Noemi Mergen, Amgen Research; Dorothea Wessiepe, Metronomia, Munich; Matthias Stelljes, University of Münster, Münster; Svenja Neumann, Heinz-August Horst, and Monika Brüggemann, University Schleswig Holstein, City Hospital, Kiel; Andreas Viardot, University of Ulm, Ulm; Reinhard Marks,
| | - Heinz-August Horst
- Max S. Topp, Hermann Einsele, and Ralf C. Bargou, Universitätsklinikum Würzburg, Würzburg; Nicola Gökbuget, Goethe University; Dieter Hoelzer, Onkologikum, Frankfurt; Gerhard Zugmaier, Petra Klappers, and Noemi Mergen, Amgen Research; Dorothea Wessiepe, Metronomia, Munich; Matthias Stelljes, University of Münster, Münster; Svenja Neumann, Heinz-August Horst, and Monika Brüggemann, University Schleswig Holstein, City Hospital, Kiel; Andreas Viardot, University of Ulm, Ulm; Reinhard Marks,
| | - Monika Brüggemann
- Max S. Topp, Hermann Einsele, and Ralf C. Bargou, Universitätsklinikum Würzburg, Würzburg; Nicola Gökbuget, Goethe University; Dieter Hoelzer, Onkologikum, Frankfurt; Gerhard Zugmaier, Petra Klappers, and Noemi Mergen, Amgen Research; Dorothea Wessiepe, Metronomia, Munich; Matthias Stelljes, University of Münster, Münster; Svenja Neumann, Heinz-August Horst, and Monika Brüggemann, University Schleswig Holstein, City Hospital, Kiel; Andreas Viardot, University of Ulm, Ulm; Reinhard Marks,
| | - Dorothea Wessiepe
- Max S. Topp, Hermann Einsele, and Ralf C. Bargou, Universitätsklinikum Würzburg, Würzburg; Nicola Gökbuget, Goethe University; Dieter Hoelzer, Onkologikum, Frankfurt; Gerhard Zugmaier, Petra Klappers, and Noemi Mergen, Amgen Research; Dorothea Wessiepe, Metronomia, Munich; Matthias Stelljes, University of Münster, Münster; Svenja Neumann, Heinz-August Horst, and Monika Brüggemann, University Schleswig Holstein, City Hospital, Kiel; Andreas Viardot, University of Ulm, Ulm; Reinhard Marks,
| | - Chris Holland
- Max S. Topp, Hermann Einsele, and Ralf C. Bargou, Universitätsklinikum Würzburg, Würzburg; Nicola Gökbuget, Goethe University; Dieter Hoelzer, Onkologikum, Frankfurt; Gerhard Zugmaier, Petra Klappers, and Noemi Mergen, Amgen Research; Dorothea Wessiepe, Metronomia, Munich; Matthias Stelljes, University of Münster, Münster; Svenja Neumann, Heinz-August Horst, and Monika Brüggemann, University Schleswig Holstein, City Hospital, Kiel; Andreas Viardot, University of Ulm, Ulm; Reinhard Marks,
| | - Shilpa Alekar
- Max S. Topp, Hermann Einsele, and Ralf C. Bargou, Universitätsklinikum Würzburg, Würzburg; Nicola Gökbuget, Goethe University; Dieter Hoelzer, Onkologikum, Frankfurt; Gerhard Zugmaier, Petra Klappers, and Noemi Mergen, Amgen Research; Dorothea Wessiepe, Metronomia, Munich; Matthias Stelljes, University of Münster, Münster; Svenja Neumann, Heinz-August Horst, and Monika Brüggemann, University Schleswig Holstein, City Hospital, Kiel; Andreas Viardot, University of Ulm, Ulm; Reinhard Marks,
| | - Noemi Mergen
- Max S. Topp, Hermann Einsele, and Ralf C. Bargou, Universitätsklinikum Würzburg, Würzburg; Nicola Gökbuget, Goethe University; Dieter Hoelzer, Onkologikum, Frankfurt; Gerhard Zugmaier, Petra Klappers, and Noemi Mergen, Amgen Research; Dorothea Wessiepe, Metronomia, Munich; Matthias Stelljes, University of Münster, Münster; Svenja Neumann, Heinz-August Horst, and Monika Brüggemann, University Schleswig Holstein, City Hospital, Kiel; Andreas Viardot, University of Ulm, Ulm; Reinhard Marks,
| | - Hermann Einsele
- Max S. Topp, Hermann Einsele, and Ralf C. Bargou, Universitätsklinikum Würzburg, Würzburg; Nicola Gökbuget, Goethe University; Dieter Hoelzer, Onkologikum, Frankfurt; Gerhard Zugmaier, Petra Klappers, and Noemi Mergen, Amgen Research; Dorothea Wessiepe, Metronomia, Munich; Matthias Stelljes, University of Münster, Münster; Svenja Neumann, Heinz-August Horst, and Monika Brüggemann, University Schleswig Holstein, City Hospital, Kiel; Andreas Viardot, University of Ulm, Ulm; Reinhard Marks,
| | - Dieter Hoelzer
- Max S. Topp, Hermann Einsele, and Ralf C. Bargou, Universitätsklinikum Würzburg, Würzburg; Nicola Gökbuget, Goethe University; Dieter Hoelzer, Onkologikum, Frankfurt; Gerhard Zugmaier, Petra Klappers, and Noemi Mergen, Amgen Research; Dorothea Wessiepe, Metronomia, Munich; Matthias Stelljes, University of Münster, Münster; Svenja Neumann, Heinz-August Horst, and Monika Brüggemann, University Schleswig Holstein, City Hospital, Kiel; Andreas Viardot, University of Ulm, Ulm; Reinhard Marks,
| | - Ralf C. Bargou
- Max S. Topp, Hermann Einsele, and Ralf C. Bargou, Universitätsklinikum Würzburg, Würzburg; Nicola Gökbuget, Goethe University; Dieter Hoelzer, Onkologikum, Frankfurt; Gerhard Zugmaier, Petra Klappers, and Noemi Mergen, Amgen Research; Dorothea Wessiepe, Metronomia, Munich; Matthias Stelljes, University of Münster, Münster; Svenja Neumann, Heinz-August Horst, and Monika Brüggemann, University Schleswig Holstein, City Hospital, Kiel; Andreas Viardot, University of Ulm, Ulm; Reinhard Marks,
| |
Collapse
|
283
|
Spinelli O, Tosi M, Guinea Montalvo ML, Peruta B, Parolini M, Scattolin AM, Maino E, Viero P, Rambaldi A, Bassan R. Prognostic impact of minimal residual disease in adult acute lymphoblastic leukemia. Int J Hematol Oncol 2014. [DOI: 10.2217/ijh.14.41] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
SUMMARY While adult acute lymphoblastic leukemia (ALL) is curable in 40–50% of the patients, the individual prognosis is rather unpredictable due to associated biological and clinical risk factors. In both B- and T-precursor ALL, minimal residual disease (MRD) represents the most sensitive prognostic marker, useful to support critical treatment decisions, ranging from allogeneic stem cell transplantation in patients with inadequate MRD response to chemotherapy only in MRD responsive ones. This optimized risk-adapted strategy allows to spare transplant-associated morbidity and mortality in patients curable by chemotherapy. Further progress is expected from the integration of the MRD-based strategy with improved pediatric-type regimens and novel targeting agents for discrete ALL subsets. These changes are increasing the cure rate to above 50%.
Collapse
Affiliation(s)
- Orietta Spinelli
- Hematology & Bone Marrow Transplant Unit of Azienda Ospedaliera Papa Giovanni XXIII, Bergamo, Italy
| | - Manuela Tosi
- Hematology & Bone Marrow Transplant Unit of Azienda Ospedaliera Papa Giovanni XXIII, Bergamo, Italy
| | | | - Barbara Peruta
- Hematology & Bone Marrow Transplant Unit of Azienda Ospedaliera Papa Giovanni XXIII, Bergamo, Italy
| | - Margherita Parolini
- Hematology & Bone Marrow Transplant Unit of Azienda Ospedaliera Papa Giovanni XXIII, Bergamo, Italy
| | - Anna Maria Scattolin
- Hematology & Bone Marrow Transplant Unit, Ospedale dell'Angelo e SS. Giovanni e Paolo, Via Paccagnella 11, 30174 Mestre-Venezia, Mestre-Venezia, Italy
| | - Elena Maino
- Hematology & Bone Marrow Transplant Unit, Ospedale dell'Angelo e SS. Giovanni e Paolo, Via Paccagnella 11, 30174 Mestre-Venezia, Mestre-Venezia, Italy
| | - Piera Viero
- Hematology & Bone Marrow Transplant Unit, Ospedale dell'Angelo e SS. Giovanni e Paolo, Via Paccagnella 11, 30174 Mestre-Venezia, Mestre-Venezia, Italy
| | - Alessandro Rambaldi
- Hematology & Bone Marrow Transplant Unit of Azienda Ospedaliera Papa Giovanni XXIII, Bergamo, Italy
| | - Renato Bassan
- Hematology & Bone Marrow Transplant Unit, Ospedale dell'Angelo e SS. Giovanni e Paolo, Via Paccagnella 11, 30174 Mestre-Venezia, Mestre-Venezia, Italy
| |
Collapse
|
284
|
Lameris R, de Bruin RCG, Schneiders FL, van Bergen en Henegouwen PMP, Verheul HMW, de Gruijl TD, van der Vliet HJ. Bispecific antibody platforms for cancer immunotherapy. Crit Rev Oncol Hematol 2014; 92:153-65. [PMID: 25195094 DOI: 10.1016/j.critrevonc.2014.08.003] [Citation(s) in RCA: 72] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2014] [Revised: 06/11/2014] [Accepted: 08/08/2014] [Indexed: 01/10/2023] Open
Abstract
Over the past decades advances in bioengineering and expanded insight in tumor immunology have resulted in the emergence of novel bispecific antibody (bsAb) constructs that are capable of redirecting immune effector cells to the tumor microenvironment. (Pre-) clinical studies of various bsAb constructs have shown impressive results in terms of immune effector cell retargeting, target dependent activation and the induction of anti-tumor responses. This review summarizes recent advances in the field of bsAb-therapy and limitations that were encountered. Furthermore, we will discuss potential future developments that can be expected to take the bsAb approach successfully forward.
Collapse
Affiliation(s)
- Roeland Lameris
- Department of Medical Oncology, VU University Medical Center, De Boelelaan 1117, 1081 HV Amsterdam, The Netherlands
| | - Renée C G de Bruin
- Department of Medical Oncology, VU University Medical Center, De Boelelaan 1117, 1081 HV Amsterdam, The Netherlands
| | - Famke L Schneiders
- Department of Medical Oncology, VU University Medical Center, De Boelelaan 1117, 1081 HV Amsterdam, The Netherlands
| | - Paul M P van Bergen en Henegouwen
- Division of Cell Biology, Department of Biology, Faculty of Science, Utrecht University, Padualaan 8, 3584 CH Utrecht, The Netherlands
| | - Henk M W Verheul
- Department of Medical Oncology, VU University Medical Center, De Boelelaan 1117, 1081 HV Amsterdam, The Netherlands
| | - Tanja D de Gruijl
- Department of Medical Oncology, VU University Medical Center, De Boelelaan 1117, 1081 HV Amsterdam, The Netherlands
| | - Hans J van der Vliet
- Department of Medical Oncology, VU University Medical Center, De Boelelaan 1117, 1081 HV Amsterdam, The Netherlands.
| |
Collapse
|
285
|
Abstract
Despite the significant advances in modern chemotherapy, it remains challenging to treat adult patients with acute lymphoblastic leukaemia (ALL). The relapse rate remains high, and the outcome at the time of relapse is dismal. Antibody-based therapies have demonstrated promising results in this patient group. Variable mechanisms have been applied to target surface antigens (CD20 [also termed MS4A1], CD22, CD52 and CD19) that are commonly expressed on malignant leukaemia cells. In this review, we will focus on the clinical application of such therapies in adult ALL, including the naked antibodies: Rituximab, Ofatumumab, Epratuzumab and Alemtuzumab; the immunotoxins: BL22 and Combotox; the immunoconjugates: inotuzumab and SAR 3419; as well as the Bi-specific T cell engaging (BiTE)-specific antibody, Blinatumomab.
Collapse
Affiliation(s)
- Jing Ai
- Department of Hematologic Oncology and Blood Disorders, Taussig Cancer Institute, Cleveland Clinic, Cleveland, OH, USA
| | | |
Collapse
|
286
|
Bispecific T-cell engagers for cancer immunotherapy. Immunol Cell Biol 2014; 93:290-6. [PMID: 25367186 DOI: 10.1038/icb.2014.93] [Citation(s) in RCA: 306] [Impact Index Per Article: 27.8] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2014] [Revised: 09/29/2014] [Accepted: 09/29/2014] [Indexed: 01/07/2023]
Abstract
Bispecific T-cell engagers (BiTEs) are a new class of immunotherapeutic molecules intended for the treatment of cancer. These molecules enhance the patient's immune response to tumors by retargeting T cells to tumor cells. BiTEs are constructed of two single-chain variable fragments (scFv) connected in tandem by a flexible linker. One scFv binds to a T-cell-specific molecule, usually CD3, whereas the second scFv binds to a tumor-associated antigen. This structure and specificity allows a BiTE to physically link a T cell to a tumor cell, ultimately stimulating T-cell activation, tumor killing and cytokine production. BiTEs have been developed, which target several tumor-associated antigens, for a variety of both hematological and solid tumors. Several BiTEs are currently in clinical trials for their therapeutic efficacy and safety. This review examines the salient structural and functional features of BiTEs, as well as the current state of their clinical and preclinical development.
Collapse
|
287
|
Parikh SA, Litzow MR. Philadelphia chromosome-negative acute lymphoblastic leukemia: therapies under development. Future Oncol 2014; 10:2201-12. [DOI: 10.2217/fon.14.81] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
ABSTRACT Although the prognosis of adults with Philadelphia chromosome-negative acute lymphoblastic leukemia (ALL) has steadily improved over the past decade, less than 50% of patients maintain their remission at 5 years. Several approaches have been explored in the past few years including: monoclonal antibodies – either ‘naked’ (rituximab) or in combination with an immunotoxin (calicheamicin or maytansin), plant toxin (ricin), or bacterial toxin (Pseudomonas or diphtheria), and a novel bispecific T-cell-engaging antibody (blinatumomab); chimeric antigen therapy using autologous T cells that target CD19-expressing ALL; and novel agents such as proteasome inhibitors, liposomal vincristine, hypomethylating agents, nelarabine and NOTCH1 inhibitors. This review summarizes treatment approaches currently under investigation for the treatment of adult Philadelphia chromosome-negative ALL.
Collapse
Affiliation(s)
- Sameer A Parikh
- Mayo Clinic, Division of Hematology, Department of Medicine, 200 First Street SW, Rochester, MN 55905, USA
| | - Mark R Litzow
- Mayo Clinic, Division of Hematology, Department of Medicine, 200 First Street SW, Rochester, MN 55905, USA
| |
Collapse
|
288
|
Role of allogeneic hematopoietic stem cell transplantation in adult patients with acute lymphoblastic leukemia. Mediterr J Hematol Infect Dis 2014; 6:e2014065. [PMID: 25408851 PMCID: PMC4235484 DOI: 10.4084/mjhid.2014.065] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2014] [Accepted: 09/11/2014] [Indexed: 12/15/2022] Open
Abstract
Adult acute lymphoblastic leukemia (ALL) is a heterogeneous disease, due to the expression of different biological and clinical risk factors, for which allogeneic stem cell transplantation (alloHSCT) is an effective consolidation therapy. The non-relapse mortality of alloHSCT remains significantly higher compared with that of conventional chemotherapy. Therefore, one of the main challenges in the care of ALL is to establish a more precise prognostic definition to select patients who could take advantage from an alloHSCT. Currently, the use of minimal residual disease following induction and early consolidation therapy has improved the prognostic accuracy in defining ALL risk class. In Philadelphia-positive ALL, the introduction of tyrosine kinase inhibitors pre and post alloHSCT appears to improve outcomes significantly and, in the absence of specially designed clinical trials, alloHSCT remains the most effective post-remission therapy. Nowadays, alloHSCT can be performed according to various modalities encompassing the use of different conditioning regimens, as well as distinct donors and stem cell source, with a significant accessibility to transplant.
Collapse
|
289
|
Podhorecka M, Markowicz J, Szymczyk A, Pawlowski J. Target Therapy in Hematological Malignances: New Monoclonal Antibodies. INTERNATIONAL SCHOLARLY RESEARCH NOTICES 2014; 2014:701493. [PMID: 27433507 PMCID: PMC4897146 DOI: 10.1155/2014/701493] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 04/10/2014] [Revised: 09/22/2014] [Accepted: 09/23/2014] [Indexed: 11/17/2022]
Abstract
Apart from radio- and chemotherapy, monoclonal antibodies (MoAbs) represent a new, more selective tool in the treatment of hematological malignancies. MoAbs bind with the specific antigens of the tumors. This interaction is a basis for targeted therapies which exhibit few side effects and significant antitumor activity. This review provides an overview of the functional characteristics of MoAbs, with some examples of their clinical application. The promising results in the treatment of hematological malignancies have led to the more frequent usage of MoAbs in the therapy. Development of MoAbs is a subject of extensive research. They are a promising method of cancer treatment in the future.
Collapse
Affiliation(s)
- Monika Podhorecka
- Department of Hematooncology and Bone Marrow Transplantation, Medical University of Lublin, Staszica 11, 20-081 Lublin, Poland
| | - Justyna Markowicz
- Students Scientific Association at the Department of Hematooncology and Bone Marrow Transplantation, Medical University of Lublin, Lublin, Poland
| | - Agnieszka Szymczyk
- Department of Hematooncology and Bone Marrow Transplantation, Medical University of Lublin, Staszica 11, 20-081 Lublin, Poland
| | - Johannes Pawlowski
- Students Scientific Association at the Department of Hematooncology and Bone Marrow Transplantation, Medical University of Lublin, Lublin, Poland
| |
Collapse
|
290
|
Vacchelli E, Eggermont A, Galon J, Sautès-Fridman C, Zitvogel L, Kroemer G, Galluzzi L. Trial watch: Monoclonal antibodies in cancer therapy. Oncoimmunology 2014; 2:e22789. [PMID: 23482847 PMCID: PMC3583934 DOI: 10.4161/onci.22789] [Citation(s) in RCA: 82] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
During the past 20 years, dozens-if not hundreds-of monoclonal antibodies have been developed and characterized for their capacity to mediate antineoplastic effects, either as they activate/enhance tumor-specific immune responses, either as they interrupt cancer cell-intrinsic signal transduction cascades, either as they specifically delivery toxins to malignant cells or as they block the tumor-stroma interaction. Such an intense research effort has lead to the approval by FDA of no less than 14 distinct molecules for use in humans affected by hematological or solid malignancies. In the inaugural issue of OncoImmunology, we briefly described the scientific rationale behind the use of monoclonal antibodies in cancer therapy and discussed recent, ongoing clinical studies investigating the safety and efficacy of this approach in patients. Here, we summarize the latest developments in this exciting area of clinical research, focusing on high impact studies that have been published during the last 15 months and clinical trials launched in the same period to investigate the therapeutic profile of promising, yet hitherto investigational, monoclonal antibodies.
Collapse
Affiliation(s)
- Erika Vacchelli
- Institut Gustave Roussy; Villejuif, France ; Université Paris-Sud/Paris XI; Le Kremlin-Bicêtre, France ; INSERM; U848; Villejuif, France
| | | | | | | | | | | | | |
Collapse
|
291
|
Maude SL, Frey N, Shaw PA, Aplenc R, Barrett DM, Bunin NJ, Chew A, Gonzalez VE, Zheng Z, Lacey SF, Mahnke YD, Melenhorst JJ, Rheingold SR, Shen A, Teachey DT, Levine BL, June CH, Porter DL, Grupp SA. Chimeric antigen receptor T cells for sustained remissions in leukemia. N Engl J Med 2014; 371:1507-17. [PMID: 25317870 PMCID: PMC4267531 DOI: 10.1056/nejmoa1407222] [Citation(s) in RCA: 4162] [Impact Index Per Article: 378.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
BACKGROUND Relapsed acute lymphoblastic leukemia (ALL) is difficult to treat despite the availability of aggressive therapies. Chimeric antigen receptor-modified T cells targeting CD19 may overcome many limitations of conventional therapies and induce remission in patients with refractory disease. METHODS We infused autologous T cells transduced with a CD19-directed chimeric antigen receptor (CTL019) lentiviral vector in patients with relapsed or refractory ALL at doses of 0.76×10(6) to 20.6×10(6) CTL019 cells per kilogram of body weight. Patients were monitored for a response, toxic effects, and the expansion and persistence of circulating CTL019 T cells. RESULTS A total of 30 children and adults received CTL019. Complete remission was achieved in 27 patients (90%), including 2 patients with blinatumomab-refractory disease and 15 who had undergone stem-cell transplantation. CTL019 cells proliferated in vivo and were detectable in the blood, bone marrow, and cerebrospinal fluid of patients who had a response. Sustained remission was achieved with a 6-month event-free survival rate of 67% (95% confidence interval [CI], 51 to 88) and an overall survival rate of 78% (95% CI, 65 to 95). At 6 months, the probability that a patient would have persistence of CTL019 was 68% (95% CI, 50 to 92) and the probability that a patient would have relapse-free B-cell aplasia was 73% (95% CI, 57 to 94). All the patients had the cytokine-release syndrome. Severe cytokine-release syndrome, which developed in 27% of the patients, was associated with a higher disease burden before infusion and was effectively treated with the anti-interleukin-6 receptor antibody tocilizumab. CONCLUSIONS Chimeric antigen receptor-modified T-cell therapy against CD19 was effective in treating relapsed and refractory ALL. CTL019 was associated with a high remission rate, even among patients for whom stem-cell transplantation had failed, and durable remissions up to 24 months were observed. (Funded by Novartis and others; CART19 ClinicalTrials.gov numbers, NCT01626495 and NCT01029366.).
Collapse
Affiliation(s)
- Shannon L Maude
- From the Division of Oncology, Children's Hospital of Philadelphia (S.L.M., R.A., D.M.B., N.J.B., S.R.R., D.T.T., S.A.G.), the Departments of Pediatrics (S.L.M., R.A., D.M.B., N.J.B., S.R.R., D.T.T., S.A.G.), Biostatistics and Epidemiology (P.A.S., R.A.), and Pathology and Laboratory Medicine (J.J.M., B.L.L., C.H.J., S.A.G.), the Division of Hematology-Oncology (N.F., D.L.P.), and Abramson Cancer Center (N.F., A.C., V.E.G., Z.Z., S.F.L., Y.D.M., J.J.M., B.L.L., C.H.J., D.L.P., S.A.G.), Perelman School of Medicine, University of Pennsylvania - all in Philadelphia; and Novartis Pharmaceuticals, East Hanover, NJ (A.S.)
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
292
|
Abstract
Pediatric acute lymphoblastic leukemia (ALL) cure rates have markedly improved over the past years to approximately 85%, but remain at 40%-50% in adults. Redefining current adult chemotherapy regimens is likely to improve the natural course of the disease, but new agents are needed. Immunotherapy approaches for pre-B ALL are in the forefront of research on novel agents; in particular, advances are being made in manipulating autologous T cells either by infusion of a bifunctional antibody (eg, blinatumomab) or by ex vivo genetic modification of chimeric antigen receptors (CARs). The natural course of Philadelphia positive ALL has already improved by targeting ABL/BCR1. Other mutated genes are being discovered and novel small molecules that target their products are being studied in clinical trials. Finally, ALL is a heterogeneous disease and novel agents are likely to impact the natural course of smaller populations of biologically defined ALL subtypes.
Collapse
Affiliation(s)
- Dan Douer
- Leukemia Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, Weill Cornell Medical College, 1275 York Ave, New York, NY 10065, USA.
| |
Collapse
|
293
|
Bleakley M, Kean L. Future of allogeneic hematopoietic stem cell transplantation for chemotherapy-resistant pediatric acute leukemia: potential advances. Int J Hematol Oncol 2014. [DOI: 10.2217/ijh.14.37] [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)
- Marie Bleakley
- Fred Hutchinson Cancer Research Center, Seattle, WA, USA
- University of Washington Department of Pediatrics, Seattle, WA, USA
| | - Leslie Kean
- Fred Hutchinson Cancer Research Center, Seattle, WA, USA
- University of Washington Department of Pediatrics, Seattle, WA, USA
- Ben Towne Center for Childhood Cancer Research, Seattle Children's Research Institute, Seattle, WA, USA
| |
Collapse
|
294
|
Henig I, Zuckerman T. Hematopoietic stem cell transplantation-50 years of evolution and future perspectives. Rambam Maimonides Med J 2014; 5:e0028. [PMID: 25386344 PMCID: PMC4222417 DOI: 10.5041/rmmj.10162] [Citation(s) in RCA: 117] [Impact Index Per Article: 10.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/11/2023] Open
Abstract
Hematopoietic stem cell transplantation is a highly specialized and unique medical procedure. Autologous transplantation allows the administration of high-dose chemotherapy without prolonged bone marrow aplasia. In allogeneic transplantation, donor-derived stem cells provide alloimmunity that enables a graft-versus-tumor effect to eradicate residual disease and prevent relapse. The first allogeneic transplantation was performed by E. Donnall Thomas in 1957. Since then the field has evolved and expanded worldwide. New indications beside acute leukemia and aplastic anemia have been constantly explored and now include congenital disorders of the hematopoietic system, metabolic disorders, and autoimmune disease. The use of matched unrelated donors, umbilical cord blood units, and partially matched related donors has dramatically extended the availability of allogeneic transplantation. Transplant-related mortality has decreased due to improved supportive care, including better strategies to prevent severe infections and with the incorporation of reduced-intensity conditioning protocols that lowered the toxicity and allowed for transplantation in older patients. However, disease relapse and graft-versus-host disease remain the two major causes of mortality with unsatisfactory progress. Intense research aiming to improve adoptive immunotherapy and increase graft-versus-leukemia response while decreasing graft-versus-host response might bring the next breakthrough in allogeneic transplantation. Strategies of graft manipulation, tumor-associated antigen vaccinations, monoclonal antibodies, and adoptive cellular immunotherapy have already proved clinically efficient. In the following years, allogeneic transplantation is likely to become more complex, more individualized, and more efficient.
Collapse
Affiliation(s)
- Israel Henig
- Department of Hematology and Bone Marrow Transplantation, Rambam Health Care Campus, Haifa, Israel
| | - Tsila Zuckerman
- 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
| |
Collapse
|
295
|
Magge RS, DeAngelis LM. The double-edged sword: Neurotoxicity of chemotherapy. Blood Rev 2014; 29:93-100. [PMID: 25445718 DOI: 10.1016/j.blre.2014.09.012] [Citation(s) in RCA: 86] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2014] [Revised: 09/17/2014] [Accepted: 09/19/2014] [Indexed: 02/06/2023]
Abstract
The number of available therapies for hematologic malignancies continues to grow at a rapid pace. Unfortunately, many of these treatments carry both central and peripheral nervous system toxicities, potentially limiting a patient's ability to tolerate a full course of treatment. Neurotoxicity with chemotherapy is common and second only to myelosuppression as a reason to limit dosing. This review addresses the neurotoxicity of newly available therapeutic agents including brentuximab vedotin and blinatumomab as well as classic ones such as methotrexate, vinca alkaloids and platinums. Although peripheral neuropathy is common with many drugs, other complications such as seizures and encephalopathy may require more immediate attention. Rapid recognition of adverse neurologic effects may lead to earlier treatment and appropriate adjustment of dosing regimens. In addition, knowledge of common toxicities may help differentiate chemotherapy-related symptoms from actual progression of cancer into the CNS.
Collapse
Affiliation(s)
- Rajiv S Magge
- Department of Neurology, Memorial Sloan Kettering Cancer Center, 1275 York Avenue, New York, NY 10065, USA.
| | - Lisa M DeAngelis
- Department of Neurology, Memorial Sloan Kettering Cancer Center, 1275 York Avenue, New York, NY 10065, USA; Department of Neurology, Weill Cornell Medical College, New York, NY 10065, USA.
| |
Collapse
|
296
|
Israyelyan A, Goldstein L, Tsai W, Aquino L, Forman SJ, Nakamura R, Diamond DJ. Real-time assessment of relapse risk based on the WT1 marker in acute leukemia and myelodysplastic syndrome patients after hematopoietic cell transplantation. Bone Marrow Transplant 2014; 50:26-33. [PMID: 25243629 PMCID: PMC4286541 DOI: 10.1038/bmt.2014.209] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2014] [Revised: 08/07/2014] [Accepted: 08/13/2014] [Indexed: 02/04/2023]
Abstract
Relapse is the major cause of treatment failure after allogeneic hematopoietic cell transplantation (alloHCT) for acute leukemia and myelodysplastic syndrome (MDS). Wilms' tumor Ag (WT1) is overexpressed in the majority of acute leukemia and MDS patients and has been proposed as a universal diagnostic marker for detection of impending relapse. Comprehensive studies have shown that WT1 transcript levels have predictive value in acute leukemia patients in CR after chemotherapy. However, the focus of this study is the period after alloHCT for predicting relapse onset. We analyzed the accumulation of WT1 mRNA transcripts in PB of 82 leukemia and MDS patients and defined specific molecular ratios for relapse prediction. The extensively validated WT1/c-ABL ratio was used to normalize increases in WT1 transcript levels. The observed lead time of crossing or exceeding set WT1 levels is presented along with linear interpolation to estimate the calculated day the WT1 thresholds were crossed. The WT1/c-ABL transcript ratio of 50 or above yielded 100% specificity and 75% sensitivity reliably predicting future relapse with an observed average of 29.4 days (s.d.=19.8) and a calculated average of 63 days (s.d.=29.3) lead time before morphologic confirmation. A lower ratio of 20 or above gave lower specificity, but higher sensitivity (84.8% and 87.5%, respectively) identified more patients who relapsed, at earlier times, providing an earlier warning with actual average lead time of 49.1 days (s.d.=30.8) and calculated average of 78 days (s.d.=28.8). WT1 transcript levels serve as a diagnostic relapse test with greater sensitivity than the morphologic approach used in the clinic as a readout.
Collapse
Affiliation(s)
- A Israyelyan
- Division of Translational Vaccine Research, Beckman Research Institute of City of Hope, Duarte, CA, USA
| | - L Goldstein
- Division of Biostatistics, Department of Information Sciences, Duarte, CA, USA
| | - W Tsai
- Department of Hematology/Hematopoietic Cell Transplantation, Duarte, CA, USA
| | - L Aquino
- Clinical Trials Office, City of Hope Comprehensive Cancer Center, Duarte, CA, USA
| | - S J Forman
- Department of Hematology/Hematopoietic Cell Transplantation, Duarte, CA, USA
| | - R Nakamura
- Department of Hematology/Hematopoietic Cell Transplantation, Duarte, CA, USA
| | - D J Diamond
- Division of Translational Vaccine Research, Beckman Research Institute of City of Hope, Duarte, CA, USA
| |
Collapse
|
297
|
Zimmerman Z, Maniar T, Nagorsen D. Unleashing the clinical power of T cells: CD19/CD3 bi-specific T cell engager (BiTE(R)) antibody construct blinatumomab as a potential therapy. Int Immunol 2014; 27:31-7. [DOI: 10.1093/intimm/dxu089] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022] Open
|
298
|
Long-term follow-up of serum immunoglobulin levels in blinatumomab-treated patients with minimal residual disease-positive B-precursor acute lymphoblastic leukemia. Blood Cancer J 2014; 4:244. [PMID: 25192414 PMCID: PMC4183773 DOI: 10.1038/bcj.2014.64] [Citation(s) in RCA: 60] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022] Open
|
299
|
Prognostic significance and treatment implications of minimal residual disease studies in Philadelphia-negative adult acute lymphoblastic leukemia. Mediterr J Hematol Infect Dis 2014; 6:e2014062. [PMID: 25237475 PMCID: PMC4165493 DOI: 10.4084/mjhid.2014.062] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2014] [Accepted: 08/22/2014] [Indexed: 11/08/2022] Open
Abstract
Acute lymphoblastic leukemia (ALL) is curable in about 40-50% of adult patients, however this is subject to ample variations owing to several host- and disease-related prognostic characteristics. Currently, the study of minimal residual disease (MRD) following induction and early consolidation therapy stands out as the most sensitive individual prognostic marker to define the risk of relapse following the achievement of remission, and ultimately that of treatment failure or success. Because substantial therapeutic advancement is now being achieved using intensified pediatric-type regimens, MRD analysis is especially useful to orientate stem cell transplantation choices. These strategic innovations are progressively leading to greater than 50% cure rates.
Collapse
|
300
|
Tsirigotis P, Shimoni A, Nagler A. The expanding horizon of immunotherapy in the treatment of malignant disorders: allogeneic hematopoietic stem cell transplantation and beyond. Ann Med 2014; 46:384-96. [PMID: 24888385 DOI: 10.3109/07853890.2014.918463] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
Allogeneic hematopoietic stem cell transplantation (allo-SCT) is a very effective therapeutic modality with curative potential in patients with hematological malignancies. The therapeutic efficacy is mainly based on the alloreactive reaction of donor lymphocytes against malignant cells of the recipient named as 'graft-versus-leukemia' or 'graft-versus-tumor' (GVL, GVT) effect. However, besides the beneficial GVL effect, alloreactive reaction attacks normal cells and provokes the deleterious 'graft-versus-host disease' (GVHD) which represents the major limitation of allo-SCT. Current trials have focused on a dual goal: augmentation of GVL and complete abolishment of GVHD. From a theoretical point of view complete dissociation of GVL from GVHD can occur by selecting antigenic targets present on malignant and absent from normal cells. Hematopoietic tissue-restricted minor histocompatibility antigens and leukemia or tumor-associated antigens are ideal candidates for tumor-targeted immunotherapy. Other options for inducing anti-tumor immunity in the absence of GVHD are natural killer (NK) cell immunotherapy, amplification of immune responses by using monoclonal antibodies, and bispecific T and NK-cell engagers. Genetically modified immune effectors such as T-cells armed with chimeric antigen receptors (CAR) or transduced with T-cell receptors with anti-tumor specificity are another exciting field of immunotherapy against malignancies.
Collapse
Affiliation(s)
- Panagiotis Tsirigotis
- Second Department of Internal Medicine, Propaedeutic, Attikon General University Hospital, University of Athens , Greece
| | | | | |
Collapse
|