401
|
Wang Y, Tschautscher MA, Rabe KG, Call TG, Leis JF, Kenderian SS, Kay NE, Muchtar E, Van Dyke DL, Koehler AB, Schwager SM, Slager SL, Parikh SA, Ding W. Clinical characteristics and outcomes of Richter transformation: experience of 204 patients from a single center. Haematologica 2019; 105:765-773. [PMID: 31197071 PMCID: PMC7049354 DOI: 10.3324/haematol.2019.224121] [Citation(s) in RCA: 76] [Impact Index Per Article: 12.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2019] [Accepted: 06/12/2019] [Indexed: 12/14/2022] Open
Abstract
The natural history, prognostication and optimal treatment of Richter transformation developed from chronic lymphocytic leukemia (CLL) are not well defined. We report the clinical characteristics and outcomes of a large series of biopsy-confirmed Richter transformation (diffuse large B-cell lymphoma or high grade B-cell lymphoma, n=204) cases diagnosed from 1993 to 2018. After a median follow up of 67.0 months, the median overall survival (OS) was 12.0 months. Patients who received no prior treatment for CLL had significantly better OS (median 46.3 vs. 7.8 months; P<0.001). Patients with elevated lactate dehydrogenase (median 6.2 vs. 39.9 months; P<0.0001) or TP53 disruption (median 8.3 vs. 12.8 months; P=0.046) had worse OS than those without. Immunoglobulin heavy chain variable region gene mutation, cell of origin, Myc/Bcl-2 double expression and MYC/BCL2/BCL6 double-/triple-hit status were not associated with OS. In multivariable Cox regression, elevated lactate dehydrogenase [Hazard ratio (HR) 2.3, 95% Confidence Interval (CI): 1.3-4.1; P=0.01], prior CLL treatment (HR 2.0, 95%CI: 1.2-3.5; P=0.01), and older age (HR 1.03, 95%CI: 1.01-1.05; P=0.01) were associated with worse OS. Twenty-four (12%) patients underwent stem cell transplant (20 autologous and 4 allogeneic), and had a median post-transplant survival of 55.4 months. In conclusion, the overall outcome of Richter transformation is poor. Richter transformation developed in patients with untreated CLL has significantly better survival. Stem cell transplant may benefit select patients.
Collapse
Affiliation(s)
- Yucai Wang
- Division of Hematology, Mayo Clinic, Rochester, MN
| | | | - Kari G Rabe
- Division of Biomedical Statistics and Informatics, Mayo Clinic, Rochester, MN
| | | | - Jose F Leis
- Division of Hematology and Medical Oncology, Mayo Clinic, Phoenix, AZ
| | | | - Neil E Kay
- Division of Hematology, Mayo Clinic, Rochester, MN
| | - Eli Muchtar
- Division of Hematology, Mayo Clinic, Rochester, MN
| | - Daniel L Van Dyke
- Division of Laboratory Genetics and Genomics, Mayo Clinic, Rochester, MN, USA
| | | | | | - Susan L Slager
- Division of Biomedical Statistics and Informatics, Mayo Clinic, Rochester, MN
| | | | - Wei Ding
- Division of Hematology, Mayo Clinic, Rochester, MN
| |
Collapse
|
402
|
Abstract
PURPOSE OF REVIEW While chemoimmunotherapy has improved outcomes in chronic lymphocytic leukemia (CLL), it is not curative, has significant relapse rates, and is not always well tolerated. Recently, novel targeted therapies have been developed to increase response rates and reduce toxicity, especially in high-risk disease. Current goals of CLL therapies are to produce deep and durable, especially minimal residual disease (MRD)-negative, remissions so as to allow patients to ultimately discontinue treatment for a while. Whether this can be achieved with single agents or combination regimens is being investigated. Here, we comment on what the results of recent and ongoing clinical trials mean for the future of CLL therapy. RECENT FINDINGS Large trials have proven the efficacy of novel therapies including small-molecule inhibitors like ibrutinib, idelalisib, and venetoclax. These agents are approved as monotherapy for first-line treatment and/or in the relapsed/refractory setting. However, it appears that combining these drugs with other novel agents or with chemoimmunotherapy can give higher rates of MRD-negative remission, and delay disease resistance. Chimeric antigen receptor-T cells may change the outlook for patients with heavily refractory CLL. Further research will determine which drug combinations are optimal for the various subgroups of CLL patients.
Collapse
|
403
|
Marei HE, Althani A, Caceci T, Arriga R, Sconocchia T, Ottaviani A, Lanzilli G, Roselli M, Caratelli S, Cenciarelli C, Sconocchia G. Recent perspective on CAR and Fcγ-CR T cell immunotherapy for cancers: Preclinical evidence versus clinical outcomes. Biochem Pharmacol 2019; 166:335-346. [PMID: 31176617 DOI: 10.1016/j.bcp.2019.06.002] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2019] [Accepted: 06/03/2019] [Indexed: 12/16/2022]
Abstract
The chimeric antigen receptor T cell (CAR-T cell) immunotherapy currently represents a hot research trend and it is expected to revolutionize the field of cancer therapy. Promising outcomes have been achieved using CAR-T cell therapy for haematological malignancies. Despite encouraging results, several challenges still pose eminent hurdles before being fully recognized. Directing CAR-T cells to target a single tumour associated antigen (TAA) as the case in haematological malignancies might be much simpler than targeting the extensive inhibitory microenvironments associated with solid tumours. This review focuses on the basic principles involved in development of CAR-T cells, emphasizing the differences between humoral IgG, T-cell receptors, CAR and Fcγ-CR constructs. It also highlights the complex inhibitory network that is usually associated with solid tumours, and tackles recent advances in the clinical studies that have provided great hope for the future use of CAR-T cell immunotherapy. While current Fcγ-CR T cell immunotherapy is in pre-clinical stage, is expected to provide a sound therapeutic approach to add to existing classical chemo- and radio-therapeutic modalities.
Collapse
Affiliation(s)
- Hany E Marei
- Department of Cytology and Histology, Faculty of Veterinary Medicine, Mansoura University, Mansoura 35116, Egypt.
| | - Asma Althani
- Biomedical Research Center, Qatar University, Doha 2713, Qatar
| | - Thomas Caceci
- Biomedical Science Education, Virginia Tech Carilion School of Medicine, Roanoke, VA, United States
| | - Roberto Arriga
- Department of Systems Medicine, Endocrinology and Medical Oncology, University of Rome "Tor Vergata", Rome, Italy
| | - Tommaso Sconocchia
- Otto Loewi Research Center, Chair of Immunology and Pathophysiology, Medical University of Graz, Graz, Austria
| | | | | | - Mario Roselli
- Department of Systems Medicine, Endocrinology and Medical Oncology, University of Rome "Tor Vergata", Rome, Italy
| | - Sara Caratelli
- Institute of Translational Pharmacology-CNR, Rome, Italy
| | | | | |
Collapse
|
404
|
Jacoby E. Relapse and Resistance to CAR-T Cells and Blinatumomab in Hematologic Malignancies. Clin Hematol Int 2019; 1:79-84. [PMID: 34595414 PMCID: PMC8432394 DOI: 10.2991/chi.d.190219.001] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2019] [Accepted: 02/08/2019] [Indexed: 01/08/2023] Open
Abstract
Application of immunotherapeutic modalities changed paradigms in the treatment of hematologic malignancies, with regards to drug manufacturing, treatment protocols, short- and long-term toxicities. FDA-approved therapies, including blinatumomab, tisagenlecleucel, and axicabtagene ciloleucel, target T cells to attack healthy and malignant cells expressing CD19, leading to high response rates in previously heavily treated patients, and to durable remissions in the absence of further therapies. Nevertheless, despite paucity of long-term data, some patients are resistant to these agents, and many relapse. This review will discuss the mechanisms of failure of these immune-based therapies, and offer guidelines to the practicing physician.
Collapse
Affiliation(s)
- Elad Jacoby
- Division of Pediatric Hematology and Oncology, The Edmond and Lily Safra Children's Hospital, Sheba Medical Center, Tel Hashomer, and Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| |
Collapse
|
405
|
Schubert ML, Schmitt A, Sellner L, Neuber B, Kunz J, Wuchter P, Kunz A, Gern U, Michels B, Hofmann S, Hückelhoven-Krauss A, Kulozik A, Ho AD, Müller-Tidow C, Dreger P, Schmitt M. Treatment of patients with relapsed or refractory CD19+ lymphoid disease with T lymphocytes transduced by RV-SFG.CD19.CD28.4-1BBzeta retroviral vector: a unicentre phase I/II clinical trial protocol. BMJ Open 2019; 9:e026644. [PMID: 31110096 PMCID: PMC6530404 DOI: 10.1136/bmjopen-2018-026644] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
INTRODUCTION Chimeric antigen receptor (CAR) T cells spark hope for patients with CD19+ B cell neoplasia, including relapsed or refractory (r/r) acute lymphoblastic leukaemia (ALL) or r/r non-Hodgkin's lymphoma (NHL). Published studies have mostly used second-generation CARs with 4-1BB or CD28 as costimulatory domains. Preclinical results of third-generation CARs incorporating both elements have shown superiority concerning longevity and proliferation. The University Hospital of Heidelberg is the first institution to run an investigator-initiated trial (IIT) CAR T cell trial (Heidelberg Chimeric Antigen Receptor T cell Trial number 1 [HD-CAR-1]) in Germany with third-generation CD19-directed CAR T cells. METHODS AND ANALYSIS Adult patients with r/r ALL (stratum I), r/r NHL including chronic lymphocytic leukaemia, diffuse large B-cell lymphoma, follicular lymphoma or mantle cell lymphoma (stratum II) as well as paediatric patients with r/r ALL (stratum III) will be treated with autologous T-lymphocytes transduced by third-generation RV-SFG.CD19.CD28.4-1BB zeta retroviral vector (CD19.CAR T cells). The main purpose of this study is to evaluate safety and feasibility of escalating CD19.CAR T cell doses (1-20×106 transduced cells/m2) after lymphodepletion with fludarabine (flu) and cyclophosphamide (cyc). Patients will be monitored for cytokine release syndrome (CRS), neurotoxicity, i.e. CAR-T-cell-related encephalopathy syndrome (CRES) and/or other toxicities (primary objectives). Secondary objectives include evaluation of in vivo function and survival of CD19.CAR T cells and assessment of CD19.CAR T cell antitumour efficacy.HD-CAR-1 as a prospective, monocentric trial aims to make CAR T cell therapy accessible to patients in Europe. Currently, HD-CAR-1 is the first and only CAR T cell IIT in Germany. A third-generation Good Manufacturing Practice (GMP) grade retroviral vector, a broad spectrum of NHL, treatment of paediatric and adult ALL patients and inclusion of patients even after allogeneic stem cell transplantation (alloSCT) make this trial unique. ETHICS AND DISSEMINATION Ethical approval and approvals from the local and federal competent authorities were granted. Trial results will be reported via peer-reviewed journals and presented at conferences and scientific meetings. TRIAL REGISTRATION NUMBER Eudra CT 2016-004808-60; NCT03676504; Pre-results.
Collapse
Affiliation(s)
- Maria-Luisa Schubert
- Department of Internal Medicine V, Heidelberg University Hospital, Heidelberg, Germany
| | - Anita Schmitt
- Department of Internal Medicine V, Heidelberg University Hospital, Heidelberg, Germany
| | - Leopold Sellner
- Department of Internal Medicine V, Heidelberg University Hospital, Heidelberg, Germany
- German Cancer Consortium (DKTK), National Centre for Tumour Diseases (NCT), Heidelberg, Germany
| | - Brigitte Neuber
- Department of Internal Medicine V, Heidelberg University Hospital, Heidelberg, Germany
| | - Joachim Kunz
- Department of Pediatric Hematology, Oncology and Immunology, Children’s Hospital, Heidelberg University Hospital, Heidelberg, Germany
| | - Patrick Wuchter
- Institute of Transfusion Medicine and Immunology, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
| | - Alexander Kunz
- Department of Internal Medicine V, Heidelberg University Hospital, Heidelberg, Germany
| | - Ulrike Gern
- Department of Internal Medicine V, Heidelberg University Hospital, Heidelberg, Germany
| | - Birgit Michels
- Department of Internal Medicine V, Heidelberg University Hospital, Heidelberg, Germany
| | - Susanne Hofmann
- Department of Internal Medicine V, Heidelberg University Hospital, Heidelberg, Germany
| | | | - Andreas Kulozik
- Department of Pediatric Hematology, Oncology and Immunology, Children’s Hospital, Heidelberg University Hospital, Heidelberg, Germany
| | - Anthony D. Ho
- Department of Internal Medicine V, Heidelberg University Hospital, Heidelberg, Germany
- German Cancer Consortium (DKTK), National Centre for Tumour Diseases (NCT), Heidelberg, Germany
| | - Carsten Müller-Tidow
- Department of Internal Medicine V, Heidelberg University Hospital, Heidelberg, Germany
- German Cancer Consortium (DKTK), National Centre for Tumour Diseases (NCT), Heidelberg, Germany
| | - Peter Dreger
- Department of Internal Medicine V, Heidelberg University Hospital, Heidelberg, Germany
- German Cancer Consortium (DKTK), National Centre for Tumour Diseases (NCT), Heidelberg, Germany
| | - Michael Schmitt
- Department of Internal Medicine V, Heidelberg University Hospital, Heidelberg, Germany
- German Cancer Consortium (DKTK), National Centre for Tumour Diseases (NCT), Heidelberg, Germany
| |
Collapse
|
406
|
Tumor-Specific Reactive Oxygen Species Accelerators Improve Chimeric Antigen Receptor T Cell Therapy in B Cell Malignancies. Int J Mol Sci 2019; 20:ijms20102469. [PMID: 31109083 PMCID: PMC6566309 DOI: 10.3390/ijms20102469] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2019] [Revised: 05/13/2019] [Accepted: 05/16/2019] [Indexed: 01/16/2023] Open
Abstract
Chimeric antigen receptor T cell (CART) therapy is currently one of the most promising treatment approaches in cancer immunotherapy. However, the immunosuppressive nature of the tumor microenvironment, in particular increased reactive oxygen species (ROS) levels, provides considerable limitations. In this study, we aimed to exploit increased ROS levels in the tumor microenvironment with prodrugs of ROS accelerators, which are specifically activated in cancer cells. Upon activation, ROS accelerators induce further generation of ROS. This leads to an accumulation of ROS in tumor cells. We hypothesized that the latter cells will be more susceptible to CARTs. CD19-specific CARTs were generated with a CD19.CAR.CD28.CD137zeta third-generation retroviral vector. Cytotoxicity was determined by chromium-51 release assay. Influence of the ROS accelerators on viability and phenotype of CARTs was determined by flow cytometry. The combination of CARTs with the ROS accelerator PipFcB significantly increased their cytotoxicity in the Burkitt lymphoma cell lines Raji and Daudi, as well as primary chronic lymphocytic leukemia cells. Exposure of CARTs to PipFcB for 48 h did not influence T cell exhaustion, viability, or T cell subpopulations. In summary, the combination of CARTs with ROS accelerators may improve adoptive immunotherapy and help to overcome tumor microenvironment-mediated treatment resistance.
Collapse
|
407
|
Stoiber S, Cadilha BL, Benmebarek MR, Lesch S, Endres S, Kobold S. Limitations in the Design of Chimeric Antigen Receptors for Cancer Therapy. Cells 2019; 8:cells8050472. [PMID: 31108883 PMCID: PMC6562702 DOI: 10.3390/cells8050472] [Citation(s) in RCA: 125] [Impact Index Per Article: 20.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2019] [Revised: 05/14/2019] [Accepted: 05/15/2019] [Indexed: 12/17/2022] Open
Abstract
Cancer therapy has entered a new era, transitioning from unspecific chemotherapeutic agents to increasingly specific immune-based therapeutic strategies. Among these, chimeric antigen receptor (CAR) T cells have shown unparalleled therapeutic potential in treating refractory hematological malignancies. In contrast, solid tumors pose a much greater challenge to CAR T cell therapy, which has yet to be overcome. As this novel therapeutic modality matures, increasing effort is being invested to determine the optimal structure and properties of CARs to facilitate the transition from empirical testing to the rational design of CAR T cells. In this review, we highlight how individual CAR domains contribute to the success and failure of this promising treatment modality and provide an insight into the most notable advances in the field of CAR T cell engineering.
Collapse
Affiliation(s)
- Stefan Stoiber
- Center of Integrated Protein Science Munich (CIPS-M) and Division of Clinical Pharmacology, Department of Medicine IV, University Hospital, Ludwig-Maximilians-Universität München, Member of the German Center for Lung Research (DZL), 80337 Munich, Germany.
| | - Bruno L Cadilha
- Center of Integrated Protein Science Munich (CIPS-M) and Division of Clinical Pharmacology, Department of Medicine IV, University Hospital, Ludwig-Maximilians-Universität München, Member of the German Center for Lung Research (DZL), 80337 Munich, Germany.
| | - Mohamed-Reda Benmebarek
- Center of Integrated Protein Science Munich (CIPS-M) and Division of Clinical Pharmacology, Department of Medicine IV, University Hospital, Ludwig-Maximilians-Universität München, Member of the German Center for Lung Research (DZL), 80337 Munich, Germany.
| | - Stefanie Lesch
- Center of Integrated Protein Science Munich (CIPS-M) and Division of Clinical Pharmacology, Department of Medicine IV, University Hospital, Ludwig-Maximilians-Universität München, Member of the German Center for Lung Research (DZL), 80337 Munich, Germany.
| | - Stefan Endres
- Center of Integrated Protein Science Munich (CIPS-M) and Division of Clinical Pharmacology, Department of Medicine IV, University Hospital, Ludwig-Maximilians-Universität München, Member of the German Center for Lung Research (DZL), 80337 Munich, Germany.
- German Center for Translational Cancer Research (DKTK), 80337 Munich, Germany.
| | - Sebastian Kobold
- Center of Integrated Protein Science Munich (CIPS-M) and Division of Clinical Pharmacology, Department of Medicine IV, University Hospital, Ludwig-Maximilians-Universität München, Member of the German Center for Lung Research (DZL), 80337 Munich, Germany.
- German Center for Translational Cancer Research (DKTK), 80337 Munich, Germany.
| |
Collapse
|
408
|
Hay KA, Turtle CJ. CD19-specific chimeric antigen receptor-modified (CAR)-T cell therapy for the treatment of chronic lymphocytic leukemia in the ibrutinib era. Immunotherapy 2019; 10:251-254. [PMID: 29421980 DOI: 10.2217/imt-2017-0162] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Affiliation(s)
- Kevin A Hay
- Fred Hutchinson Cancer Research Center, Seattle, WA 98109, USA.,Department of Medicine, University of British Columbia, Vancouver, BC, V5Z 1M9, Canada
| | - Cameron J Turtle
- Fred Hutchinson Cancer Research Center, Seattle, WA 98109, USA.,Department of Medicine, University of Washington, Seattle, WA 98195, USA
| |
Collapse
|
409
|
Susanibar Adaniya SP, Cohen AD, Garfall AL. Chimeric antigen receptor T cell immunotherapy for multiple myeloma: A review of current data and potential clinical applications. Am J Hematol 2019; 94:S28-S33. [PMID: 30730071 DOI: 10.1002/ajh.25428] [Citation(s) in RCA: 31] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2019] [Revised: 01/28/2019] [Accepted: 02/04/2019] [Indexed: 12/28/2022]
Abstract
Multiple myeloma (MM) is a malignant plasma cell disorder that remains incurable for most patients despite significant improvements achieved with modern therapy. Tumor evasion is a key process in the pathogenesis of MM and a compromised immune system is associated with more aggressive forms of the disease. In contrast, the emergence of myeloma-specific immune responses after both autologous and allogeneic stem cell transplantation is associated with better prognosis. Adoptive T cell therapies may improve specific anti-myeloma immunity resulting in long-lasting remissions. CAR T cell therapies for MM are at an early stage of clinical development. To date, anti-BCMA CAR T cells have shown the greatest results in early-phase clinical trials. Toxicities have included cytokine release syndrome (CRS) and neurotoxicity. Current areas of research in CAR T cell therapies include the use of gene-editing to enhance their effectiveness and safety, the integration of CAR T cells with other therapies (immunomodulatory drugs, checkpoint inhibitors) and CAR T cells to target multiple antigens.
Collapse
Affiliation(s)
| | - Adam D. Cohen
- Abramson Cancer CenterUniversity of Pennsylvania Philadelphia Pennsylvania
| | - Alfred L. Garfall
- Abramson Cancer CenterUniversity of Pennsylvania Philadelphia Pennsylvania
| |
Collapse
|
410
|
|
411
|
Hirayama AV, Turtle CJ. Toxicities of CD19 CAR-T cell immunotherapy. Am J Hematol 2019; 94:S42-S49. [PMID: 30784102 DOI: 10.1002/ajh.25445] [Citation(s) in RCA: 100] [Impact Index Per Article: 16.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2018] [Revised: 02/18/2019] [Accepted: 02/19/2019] [Indexed: 12/30/2022]
Abstract
CD19-targeted chimeric antigen receptor (CAR)-modified T (CAR-T) cell immunotherapy has demonstrated impressive results in B-cell malignancies, and CAR-T cell therapies targeting other antigens are in development for other cancers. Cytokine release syndrome (CRS) and neurotoxicity can be life-threatening in a subset of patients. The severity of CRS and neurotoxicity can be impacted by the disease burden, lymphodepletion regimen, and CAR-T cell dose. Tocilizumab and corticosteroids have been used to manage these toxicities, enabling CD19 CAR-T cells to be administered without obvious compromise in efficacy. Consensus criteria for grading and managing toxicities will facilitate the widespread application of this treatment modality.
Collapse
Affiliation(s)
- Alexandre V. Hirayama
- Clinical Research Division and Integrated Immunotherapy Research CenterFred Hutchinson Cancer Research Center Seattle Washington
| | - Cameron J. Turtle
- Clinical Research Division and Integrated Immunotherapy Research CenterFred Hutchinson Cancer Research Center Seattle Washington
- Department of MedicineUniversity of Washington Seattle Washington
| |
Collapse
|
412
|
Bair SM, Porter DL. Accelerating chimeric antigen receptor therapy in chronic lymphocytic leukemia: The development and challenges of chimeric antigen receptor T-cell therapy for chronic lymphocytic leukemia. Am J Hematol 2019; 94:S10-S17. [PMID: 30861173 DOI: 10.1002/ajh.25457] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2019] [Accepted: 03/07/2019] [Indexed: 12/27/2022]
Abstract
Studies of chimeric antigen receptor (CAR) T-cell therapy in chronic lymphocytic leukemia (CLL) have demonstrated the potential to produce deep remissions-and possibly cures-in some patients with heavily pretreated, high-risk, relapsed, and refractory disease. Unfortunately, most clinical trials of CAR T cells in CLL report complete responses only in the minority of patients, although recent studies have begun to elucidate the factors most predictive of response. These studies have suggested strategies for optimizing CAR T-cell fitness as well as the pre-existing host immune response, approaches that will likely lead to improvements in the efficacy of CAR T cells in CLL. Treating patients earlier in the course of their disease or using combination therapies with CAR T cells may further enhance efficacy. In this review, we summarize the existing literature on CAR T cell therapy in CLL, discuss mechanisms of response and resistance, and describe challenges facing the field.
Collapse
Affiliation(s)
- Steven M. Bair
- Cell Therapy and Transplant Program, Division of Hematology‐Oncology, Perelman School of Medicine and Abramson Cancer CenterUniversity of Pennsylvania Philadelphia Pennsylvania
| | - David L. Porter
- Cell Therapy and Transplant Program, Division of Hematology‐Oncology, Perelman School of Medicine and Abramson Cancer CenterUniversity of Pennsylvania Philadelphia Pennsylvania
| |
Collapse
|
413
|
Abstract
Genetically engineered T cells are powerful new medicines, offering hope for curative responses in patients with cancer. Chimeric antigen receptor (CAR) T cells were recently approved by the US Food and Drug Administration and are poised to enter the practice of medicine for leukemia and lymphoma, demonstrating that engineered immune cells can serve as a powerful new class of cancer therapeutics. The emergence of synthetic biology approaches for cellular engineering provides a broadly expanded set of tools for programming immune cells for enhanced function. Advances in T cell engineering, genetic editing, the selection of optimal lymphocytes, and cell manufacturing have the potential to broaden T cell-based therapies and foster new applications beyond oncology, in infectious diseases, organ transplantation, and autoimmunity.
Collapse
Affiliation(s)
- Sonia Guedan
- Department of Hematology, Hospital Clinic, Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), 08036 Barcelona, Spain;
- Center for Cellular Immunotherapies, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, Pennsylvania 19104, USA;
| | - Marco Ruella
- Center for Cellular Immunotherapies, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, Pennsylvania 19104, USA;
- Department of Medicine, Division of Hematology and Oncology, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, Pennsylvania 19104, USA
- Parker Institute for Cellular Immunotherapy at the University of Pennsylvania, Philadelphia, Pennsylvania 19104, USA
| | - Carl H June
- Center for Cellular Immunotherapies, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, Pennsylvania 19104, USA;
- Department of Medicine, Division of Hematology and Oncology, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, Pennsylvania 19104, USA
- Parker Institute for Cellular Immunotherapy at the University of Pennsylvania, Philadelphia, Pennsylvania 19104, USA
- Department of Pathology and Laboratory Medicine, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, Pennsylvania 19104, USA
| |
Collapse
|
414
|
Exploratory trial of a biepitopic CAR T-targeting B cell maturation antigen in relapsed/refractory multiple myeloma. Proc Natl Acad Sci U S A 2019; 116:9543-9551. [PMID: 30988175 DOI: 10.1073/pnas.1819745116] [Citation(s) in RCA: 293] [Impact Index Per Article: 48.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
Relapsed and refractory (R/R) multiple myeloma (MM) patients have very poor prognosis. Chimeric antigen receptor modified T (CAR T) cells is an emerging approach in treating hematopoietic malignancies. Here we conducted the clinical trial of a biepitope-targeting CAR T against B cell maturation antigen (BCMA) (LCAR-B38M) in 17 R/R MM cases. CAR T cells were i.v. infused after lymphodepleting chemotherapy. Two delivery methods, three infusions versus one infusion of the total CAR T dose, were tested in, respectively, 8 and 9 cases. No response differences were noted among the two delivery subgroups. Together, after CAR T cell infusion, 10 cases experienced a mild cytokine release syndrome (CRS), 6 had severe but manageable CRS, and 1 died of a very severe toxic reaction. The abundance of BCMA and cytogenetic marker del(17p) and the elevation of IL-6 were the key indicators for severe CRS. Among 17 cases, the overall response rate was 88.2%, with 13 achieving stringent complete response (sCR) and 2 reaching very good partial response (VGPR), while 1 was a nonresponder. With a median follow-up of 417 days, 8 patients remained in sCR or VGPR, whereas 6 relapsed after sCR and 1 had progressive disease (PD) after VGPR. CAR T cells were high in most cases with stable response but low in 6 out of 7 relapse/PD cases. Notably, positive anti-CAR antibody constituted a high-risk factor for relapse/PD, and patients who received prior autologous hematopoietic stem cell transplantation had more durable response. Thus, biepitopic CAR T against BCMA represents a promising therapy for R/R MM, while most adverse effects are clinically manageable.
Collapse
|
415
|
Geyer MB, Rivière I, Sénéchal B, Wang X, Wang Y, Purdon TJ, Hsu M, Devlin SM, Palomba ML, Halton E, Bernal Y, van Leeuwen DG, Sadelain M, Park JH, Brentjens RJ. Safety and tolerability of conditioning chemotherapy followed by CD19-targeted CAR T cells for relapsed/refractory CLL. JCI Insight 2019; 5:122627. [PMID: 30938714 DOI: 10.1172/jci.insight.122627] [Citation(s) in RCA: 64] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022] Open
Abstract
BACKGROUND Subgroups of patients with relapsed or refractory (R/R) chronic lymphocytic leukemia (CLL) exhibit suboptimal outcomes after standard therapies, including oral kinase inhibitors. We and others have previously reported on safety and efficacy of autologous CD19-targeted CAR T-cells for these patients; here we report safety and long-term follow-up of CAR T-cell therapy with or without conditioning chemotherapy for patients with R/R CLL and indolent B-cell non-Hodgkin lymphoma (B-NHL). METHODS We conducted a phase 1 clinical trial investigating CD19-targeted CAR T-cells incorporating a CD28 costimulatory domain (19-28z). Seventeen of 20 patients received conditioning chemotherapy prior to CAR T-cell infusion. Five patients with CLL received ibrutinib at the time of autologous T-cell collection and/or CAR T-cell administration. RESULTS This analysis included 16 patients with R/R CLL and 4 patients with R/R indolent B-NHL. Cytokine release syndrome (CRS) was observed in all 20 patients but grades 3 and 4 CRS and neurological events were uncommon (10% for each). Ex vivo expansion of T-cells and proportions of CD4+/CD8+ CAR T-cells with CD62L+CD127+ immunophenotype were significantly greater in patients on ibrutinib at leukapheresis. Three of 12 evaluable CLL patients receiving conditioning chemotherapy achieved CR (two had minimal residual disease-negative CR). All patients achieving CR remained progression-free at median follow-up of 53 months. CONCLUSION Conditioning chemotherapy and 19-28z CAR T-cells were acceptably tolerated across investigated dose levels in heavily pretreated patients with R/R CLL and indolent B-NHL, and a subgroup of patients achieved durable CR. Ibrutinib therapy may modulate autologous T-cell phenotype. TRIAL REGISTRATION ClinicalTrials.gov NCT00466531. FUNDING Juno Therapeutics.
Collapse
Affiliation(s)
- Mark B Geyer
- Department of Medicine.,Center for Cell Engineering
| | - Isabelle Rivière
- Center for Cell Engineering.,Michael G. Harris Cell Therapy and Cell Engineering Facility.,Molecular Pharmacology and Chemistry Program, and
| | - Brigitte Sénéchal
- Michael G. Harris Cell Therapy and Cell Engineering Facility.,Molecular Pharmacology and Chemistry Program, and
| | - Xiuyan Wang
- Center for Cell Engineering.,Michael G. Harris Cell Therapy and Cell Engineering Facility.,Molecular Pharmacology and Chemistry Program, and
| | - Yongzeng Wang
- Michael G. Harris Cell Therapy and Cell Engineering Facility
| | | | - Meier Hsu
- Biostatistics, Memorial Sloan Kettering Cancer Center, New York, New York, USA
| | - Sean M Devlin
- Biostatistics, Memorial Sloan Kettering Cancer Center, New York, New York, USA
| | | | | | | | | | | | - Jae H Park
- Department of Medicine.,Center for Cell Engineering
| | - Renier J Brentjens
- Department of Medicine.,Center for Cell Engineering.,Molecular Pharmacology and Chemistry Program, and
| |
Collapse
|
416
|
Yosifov DY, Wolf C, Stilgenbauer S, Mertens D. From Biology to Therapy: The CLL Success Story. Hemasphere 2019; 3:e175. [PMID: 31723816 PMCID: PMC6746030 DOI: 10.1097/hs9.0000000000000175] [Citation(s) in RCA: 50] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2018] [Revised: 12/26/2018] [Accepted: 12/28/2018] [Indexed: 11/27/2022] Open
Abstract
Chemoimmunotherapy has been the standard of care for patients with chronic lymphocytic leukemia (CLL) over the last decade. Advances in monoclonal antibody technology have resulted in the development of newer generations of anti-CD20 antibodies with improved therapeutic effectiveness. In parallel, our knowledge about the distinctive biological characteristics of CLL has progressively deepened and has revealed the importance of B-cell receptor (BCR) signaling and upregulated antiapoptotic proteins for survival and expansion of malignant cell clones. This knowledge provided the basis for development of novel targeted agents that revolutionized treatment of CLL. Ibrutinib and idelalisib inhibit the Bruton tyrosine kinase (BTK) and phosphoinositide 3-kinase (PI3K) delta, respectively, thus interfering with supportive signals coming from the microenvironment via the BCR. These drugs induce egress of CLL cells from secondary lymphoid organs and remarkably improve clinical outcomes, especially for patients with unmutated immunoglobulin heavy-chain genes or with p53 abnormalities that do not benefit from classical treatment schemes. Latest clinical trial results have established ibrutinib with or without anti-CD20 antibodies as the preferred first-line treatment for most CLL patients, which will reduce the use of chemoimmunotherapy in the imminent future. Further advances are achieved with venetoclax, a BH3-mimetic that specifically inhibits the antiapoptotic B-cell lymphoma 2 protein and thus causes rapid apoptosis of CLL cells, which translates into deep and prolonged clinical responses including high rates of minimal residual disease negativity. This review summarizes recent advances in the development of targeted CLL therapies, including new combination schemes, novel BTK and PI3K inhibitors, spleen tyrosine kinase inhibitors, immunomodulatory drugs, and cellular immunotherapy.
Collapse
Affiliation(s)
- Deyan Y. Yosifov
- Department of Internal Medicine III, Ulm University, Ulm, Germany
- Cooperation Unit “Mechanisms of Leukemogenesis”, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Christine Wolf
- Department of Internal Medicine III, Ulm University, Ulm, Germany
- Cooperation Unit “Mechanisms of Leukemogenesis”, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Stephan Stilgenbauer
- Department of Internal Medicine III, Ulm University, Ulm, Germany
- Klinik für Innere Medizin I, Universitätsklinikum des Saarlandes, Homburg, Germany
| | - Daniel Mertens
- Department of Internal Medicine III, Ulm University, Ulm, Germany
- Cooperation Unit “Mechanisms of Leukemogenesis”, German Cancer Research Center (DKFZ), Heidelberg, Germany
| |
Collapse
|
417
|
Garcia Borrega J, Gödel P, Rüger MA, Onur ÖA, Shimabukuro-Vornhagen A, Kochanek M, Böll B. In the Eye of the Storm: Immune-mediated Toxicities Associated With CAR-T Cell Therapy. Hemasphere 2019; 3:e191. [PMID: 31723828 PMCID: PMC6746039 DOI: 10.1097/hs9.0000000000000191] [Citation(s) in RCA: 62] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2018] [Revised: 02/15/2019] [Accepted: 02/15/2019] [Indexed: 12/16/2022] Open
Abstract
The success of chimeric antigen receptor (CAR)-T cell therapy with impressive response rates in hematologic malignancies but also promising data in solid tumors came along with the cognition of unexpected, potentially life-threatening immune-mediated toxicities, namely the cytokine release syndrome (CRS) and neurotoxicity recently referred to as "immune effector cell-associated neurotoxicity syndrome" (ICANS). These toxicities require urgent diagnostic and therapeutic interventions and targeted modulation of key cytokine pathways represents the mainstay of CRS treatment. However, as the underlying mechanisms of ICANS are not well understood, treatment options remain limited and further investigation is warranted. Importantly, after the recent market approval of 2 CAR-T cell constructs, the application of CAR-T cells will expand to nonacademic centers with limited experience in the management of CAR-T cell-associated toxicities. Here, we review the current evidence of CRS and ICANS pathophysiology, diagnostics, and treatment.
Collapse
Affiliation(s)
- Jorge Garcia Borrega
- Department I of Internal Medicine, Hematology-Oncology, Center of Integrated Oncology Cologne-Bonn, University Hospital of Cologne, Cologne, Germany
- Intensive Care Program, Department I of Internal Medicine, University Hospital of Cologne, Cologne, Germany
| | - Philipp Gödel
- Department I of Internal Medicine, Hematology-Oncology, Center of Integrated Oncology Cologne-Bonn, University Hospital of Cologne, Cologne, Germany
- Intensive Care Program, Department I of Internal Medicine, University Hospital of Cologne, Cologne, Germany
- Cologne Translational Immunology, University Hospital of Cologne, Cologne, Germany
| | - Maria Adele Rüger
- Department of Neurology, University Hospital of Cologne, Cologne, Germany
| | - Özgür A. Onur
- Department of Neurology, University Hospital of Cologne, Cologne, Germany
| | - Alexander Shimabukuro-Vornhagen
- Department I of Internal Medicine, Hematology-Oncology, Center of Integrated Oncology Cologne-Bonn, University Hospital of Cologne, Cologne, Germany
- Intensive Care Program, Department I of Internal Medicine, University Hospital of Cologne, Cologne, Germany
- Intensive Care in HematoOncologic Patients (iCHOP) Collaborative Group
| | - Matthias Kochanek
- Department I of Internal Medicine, Hematology-Oncology, Center of Integrated Oncology Cologne-Bonn, University Hospital of Cologne, Cologne, Germany
- Intensive Care Program, Department I of Internal Medicine, University Hospital of Cologne, Cologne, Germany
- Intensive Care in HematoOncologic Patients (iCHOP) Collaborative Group
| | - Boris Böll
- Department I of Internal Medicine, Hematology-Oncology, Center of Integrated Oncology Cologne-Bonn, University Hospital of Cologne, Cologne, Germany
- Intensive Care Program, Department I of Internal Medicine, University Hospital of Cologne, Cologne, Germany
- Intensive Care in HematoOncologic Patients (iCHOP) Collaborative Group
| |
Collapse
|
418
|
Xia AL, Xu Y, Lu XJ. Functional genomics in the era of cancer immunotherapy: challenges and clinical implications. Brief Funct Genomics 2019; 18:83-85. [PMID: 30901384 DOI: 10.1093/bfgp/elz003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2018] [Revised: 10/03/2018] [Accepted: 10/19/2018] [Indexed: 11/12/2022] Open
Affiliation(s)
- An-Liang Xia
- Department of General Surgery, Liver Transplantation Center, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Yong Xu
- Department of Nephrology, Huai'an Second People's Hospital and The Affiliated Huai'an Hospital of Xuzhou Medical University, Huai'an, China
| | - Xiao-Jie Lu
- Department of General Surgery, Liver Transplantation Center, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| |
Collapse
|
419
|
Rubin DB, Danish HH, Ali AB, Li K, LaRose S, Monk AD, Cote DJ, Spendley L, Kim AH, Robertson MS, Torre M, Smith TR, Izzy S, Jacobson CA, Lee JW, Vaitkevicius H. Neurological toxicities associated with chimeric antigen receptor T-cell therapy. Brain 2019; 142:1334-1348. [DOI: 10.1093/brain/awz053] [Citation(s) in RCA: 112] [Impact Index Per Article: 18.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2018] [Revised: 12/21/2018] [Accepted: 01/18/2019] [Indexed: 12/13/2022] Open
Affiliation(s)
- Daniel B Rubin
- Department of Neurology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
- Department of Neurology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Husain H Danish
- Department of Neurology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
- Department of Neurology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Ali Basil Ali
- Department of Neurology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Karen Li
- Department of Neurology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Sarah LaRose
- Department of Neurology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Andrew D Monk
- Department of Neurology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - David J Cote
- Department of Neurosurgery; Channing Division of Network Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA, USA
| | - Lauren Spendley
- Department of Medical Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA, USA
| | - Angela H Kim
- Department of Neurology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Matthew S Robertson
- Division of Nuclear Medicine, Department of Radiology, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA, USA
| | - Matthew Torre
- Department of Pathology, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA, USA
| | - Timothy R Smith
- Department of Neurosurgery, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA, USA
| | - Saef Izzy
- Department of Neurology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Caron A Jacobson
- Department of Medical Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA, USA
| | - Jong Woo Lee
- Department of Neurology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Henrikas Vaitkevicius
- Department of Neurology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| |
Collapse
|
420
|
Acharya UH, Dhawale T, Yun S, Jacobson CA, Chavez JC, Ramos JD, Appelbaum J, Maloney DG. Management of cytokine release syndrome and neurotoxicity in chimeric antigen receptor (CAR) T cell therapy. Expert Rev Hematol 2019; 12:195-205. [PMID: 30793644 DOI: 10.1080/17474086.2019.1585238] [Citation(s) in RCA: 41] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
INTRODUCTION Chimeric antigen receptor (CAR) T cell immunotherapy has demonstrated remarkable anti-tumor activity in B-cell malignancies and is under investigation in other hematologic malignancies and solid tumors. While highly efficacious, post-infusion T cell activity often results in massive cytokine release precipitating cytokine release syndrome (CRS), the signature toxicity of CAR T cells. This toxicity is characterized by systemic immune activation resulting in fever, hypotension, respiratory insufficiency and capillary leak. Either in conjunction with or in the absence of CRS, a subset of patients may also develop mild to severe neurotoxicity. Although the precise pathogenesis of CRS and neurotoxicity aren't fully elucidated, risk factors and mitigation strategies have been reported. Areas covered: This manuscript provides an in-depth overview of the pathogenesis, clinical characteristics, current toxicity management strategies, and future perspectives pertaining to CRS and neurotoxicity. Expert Opinion: As CAR T cell based therapies gain popularity in the management of various malignancies, the complimentary toxicities of CRS and neurotoxicity pose a clinical challenge in practice. Risk adaptive modeling incorporating disease profile, patient demographics, lymphodepletion, cell dosing, CAR T construct, and potentially cytokine gene polymorphisms may be instructive to assess individualized risk and optimal CRS/neurotoxicity management.
Collapse
Affiliation(s)
- Utkarsh H Acharya
- a Division of Medical Oncology, Department of Internal Medicine , University of Washington School of Medicine , Seattle , WA , USA.,b Clinical Research Division , Fred Hutchinson Cancer Research Center , Seattle , WA , USA.,c Divisions of Hematologic Malignancies & Immune Effector Cell Therapy, Department of Medical Oncology , Dana-Farber Cancer Institute , Boston , MA , USA
| | - Tejaswini Dhawale
- d Division of Hematology, Department of Medicine , University of Washington , Seattle , WA , USA
| | - Seongseok Yun
- e Department of Malignant Hematology , H. Lee Moffitt Cancer Center , Tampa , FL , USA
| | - Caron A Jacobson
- c Divisions of Hematologic Malignancies & Immune Effector Cell Therapy, Department of Medical Oncology , Dana-Farber Cancer Institute , Boston , MA , USA
| | - Julio C Chavez
- e Department of Malignant Hematology , H. Lee Moffitt Cancer Center , Tampa , FL , USA
| | - Jorge D Ramos
- a Division of Medical Oncology, Department of Internal Medicine , University of Washington School of Medicine , Seattle , WA , USA.,b Clinical Research Division , Fred Hutchinson Cancer Research Center , Seattle , WA , USA
| | - Jacob Appelbaum
- a Division of Medical Oncology, Department of Internal Medicine , University of Washington School of Medicine , Seattle , WA , USA.,d Division of Hematology, Department of Medicine , University of Washington , Seattle , WA , USA
| | - David G Maloney
- a Division of Medical Oncology, Department of Internal Medicine , University of Washington School of Medicine , Seattle , WA , USA.,b Clinical Research Division , Fred Hutchinson Cancer Research Center , Seattle , WA , USA
| |
Collapse
|
421
|
Liu Y, Zhao C, Gao L, Yang H, He R, Gao C. Considerations for Clinical Review of Cellular Therapy Products: Perspectives of the China Food and Drug Administration Center for Drug Evaluation. Hum Gene Ther 2019; 29:121-127. [PMID: 29446998 DOI: 10.1089/hum.2017.216] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
With increasing numbers of technical developments and clinical studies, pioneering cellular/gene therapies are now available that could cure life-threatening disease. Cellular/gene therapy products are broad-ranging and complicated, and thereby bring challenges for clinical review by regulatory agencies. This review discusses principles for the clinical review of cellular therapy products, including protection of clinical trial populations, pharmacodynamics, pharmacokinetics, dose evaluation, clinical efficacy, clinical safety, and risk-management plans. Based on these principles, key points in the clinical review of chimeric antigen receptor T-cell therapy are also discussed.
Collapse
Affiliation(s)
- Yantong Liu
- 1 Office of Clinical Evaluation of Biological Products, Center for Drug Evaluation , China Food and Drug Administration, Beijing, China
| | - Chenyang Zhao
- 1 Office of Clinical Evaluation of Biological Products, Center for Drug Evaluation , China Food and Drug Administration, Beijing, China
| | - Liucun Gao
- 1 Office of Clinical Evaluation of Biological Products, Center for Drug Evaluation , China Food and Drug Administration, Beijing, China
| | - Huan Yang
- 1 Office of Clinical Evaluation of Biological Products, Center for Drug Evaluation , China Food and Drug Administration, Beijing, China
| | - Ruyi He
- 2 Center for Drug Evaluation , China Food and Drug Administration, Beijing, China
| | - Chenyan Gao
- 1 Office of Clinical Evaluation of Biological Products, Center for Drug Evaluation , China Food and Drug Administration, Beijing, China
| |
Collapse
|
422
|
El-Daly SM, Hussein J. Genetically engineered CAR T-immune cells for cancer therapy: recent clinical developments, challenges, and future directions. J Appl Biomed 2019; 17:11. [PMID: 34907752 DOI: 10.32725/jab.2019.005] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2018] [Accepted: 01/29/2019] [Indexed: 12/27/2022] Open
Abstract
Cancer immunotherapy offers tremendous clinical outcomes in cancer management with the potential to induce sustained remission in patients with refractory disease. One of these immunotherapy modalities is the adoptive transfer of autologous T-cells that are genetically engineered ex vivo to express chimeric antigen receptors (CARs). These receptors can direct T-cells to the surface antigens of tumor cells to initiate an efficient and specific cytotoxic response against tumor cells. This review elucidates the structural features of CAR T-cells and their different generations reaching the recent 4th generation (TRUCK). The step-wise treatment process using CAR T-cell therapy and some of the updated prominent clinical applications of this treatment modality in both hematologic and solid malignancies are also covered in the present review. The success of CAR T-cell therapy is still encountered by several limitations for a widespread clinical application of this treatment modality, these challenges along with the recent innovative strategies that have been developed to overcome such drawbacks, as well as, the approaches and future directions aiming for a commercial low cost CAR T-cell immunotherapy modality, are all covered in the present review.
Collapse
Affiliation(s)
- Sherien M El-Daly
- National Research Centre, Medical Research Division, Department of Medical Biochemistry, Dokki, Cairo, Egypt.,National Research Centre, Centre of Excellence for Advanced Sciences, Cancer Biology and Genetics Laboratory, Dokki, Cairo, Egypt
| | - Jihan Hussein
- National Research Centre, Medical Research Division, Department of Medical Biochemistry, Dokki, Cairo, Egypt
| |
Collapse
|
423
|
Good Z, Borges L, Vivanco Gonzalez N, Sahaf B, Samusik N, Tibshirani R, Nolan GP, Bendall SC. Proliferation tracing with single-cell mass cytometry optimizes generation of stem cell memory-like T cells. Nat Biotechnol 2019; 37:259-266. [PMID: 30742126 PMCID: PMC6521980 DOI: 10.1038/s41587-019-0033-2] [Citation(s) in RCA: 37] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2017] [Accepted: 12/21/2018] [Indexed: 01/12/2023]
Abstract
Selective differentiation of naive T cells into multipotent T cells is of great interest clinically for the generation of cell-based cancer immunotherapies. Cellular differentiation depends crucially on division state and time. Here we adapt a dye dilution assay for tracking cell proliferative history through mass cytometry and uncouple division, time and regulatory protein expression in single naive human T cells during their activation and expansion in a complex ex vivo milieu. Using 23 markers, we defined groups of proteins controlled predominantly by division state or time and found that undivided cells account for the majority of phenotypic diversity. We next built a map of cell state changes during naive T-cell expansion. By examining cell signaling on this map, we rationally selected ibrutinib, a BTK and ITK inhibitor, and administered it before T cell activation to direct differentiation toward a T stem cell memory (TSCM)-like phenotype. This method for tracing cell fate across division states and time can be broadly applied for directing cellular differentiation.
Collapse
Affiliation(s)
- Zinaida Good
- PhD Program in Immunology, Stanford University, Stanford, CA, USA
- Baxter Laboratory in Stem Cell Biology, Stanford University, Stanford, CA, USA
- Department of Microbiology and Immunology, Stanford University, Stanford, CA, USA
- Department of Pathology, Stanford University, Stanford, CA, USA
- Parker Institute for Cancer Immunotherapy, San Francisco, CA, USA
| | - Luciene Borges
- Department of Pathology, Stanford University, Stanford, CA, USA
| | - Nora Vivanco Gonzalez
- PhD Program in Immunology, Stanford University, Stanford, CA, USA
- Department of Pathology, Stanford University, Stanford, CA, USA
| | - Bita Sahaf
- Cancer Institute, Department of Medicine, Stanford University, Stanford, CA, USA
| | - Nikolay Samusik
- Baxter Laboratory in Stem Cell Biology, Stanford University, Stanford, CA, USA
- Department of Microbiology and Immunology, Stanford University, Stanford, CA, USA
| | - Robert Tibshirani
- Department of Statistics, Stanford University, Stanford, CA, USA
- Department of Health Research and Policy, Stanford University, Stanford, CA, USA
| | - Garry P Nolan
- Baxter Laboratory in Stem Cell Biology, Stanford University, Stanford, CA, USA
- Department of Microbiology and Immunology, Stanford University, Stanford, CA, USA
- Parker Institute for Cancer Immunotherapy, San Francisco, CA, USA
| | - Sean C Bendall
- Department of Pathology, Stanford University, Stanford, CA, USA.
- Parker Institute for Cancer Immunotherapy, San Francisco, CA, USA.
| |
Collapse
|
424
|
The response to lymphodepletion impacts PFS in patients with aggressive non-Hodgkin lymphoma treated with CD19 CAR T cells. Blood 2019; 133:1876-1887. [PMID: 30782611 DOI: 10.1182/blood-2018-11-887067] [Citation(s) in RCA: 236] [Impact Index Per Article: 39.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2018] [Accepted: 02/12/2019] [Indexed: 12/21/2022] Open
Abstract
Factors associated with durable remission after CD19 chimeric antigen receptor (CAR)-modified T-cell immunotherapy for aggressive B-cell non-Hodgkin lymphoma (NHL) have not been identified. We report multivariable analyses of factors affecting response and progression-free survival (PFS) in patients with aggressive NHL treated with cyclophosphamide and fludarabine lymphodepletion followed by 2 × 106 CD19-directed CAR T cells/kg. The best overall response rate was 51%, with 40% of patients achieving complete remission. The median PFS of patients with aggressive NHL who achieved complete remission was 20.0 months (median follow-up, 26.9 months). Multivariable analysis of clinical and treatment characteristics, serum biomarkers, and CAR T-cell manufacturing and pharmacokinetic data showed that a lower pre-lymphodepletion serum lactate dehydrogenase (LDH) level and a favorable cytokine profile, defined as serum day 0 monocyte chemoattractant protein-1 (MCP-1) and peak interleukin-7 (IL-7) concentrations above the median, were associated with better PFS. MCP-1 and IL-7 concentrations increased after lymphodepletion, and higher intensity of cyclophosphamide and fludarabine lymphodepletion was associated with higher probability of a favorable cytokine profile. PFS was superior in patients who received high-intensity lymphodepletion and achieved a favorable cytokine profile compared with those who received the same intensity of lymphodepletion without achieving a favorable cytokine profile. Even in high-risk patients with pre-lymphodepletion serum LDH levels above normal, a favorable cytokine profile after lymphodepletion was associated with a low risk of a PFS event. Strategies to augment the cytokine response to lymphodepletion could be tested in future studies of CD19 CAR T-cell immunotherapy for aggressive B-cell NHL. This trial was registered at www.clinicaltrials.gov as #NCT01865617.
Collapse
|
425
|
Chimeric Antigen Receptor T Cells Targeting CD19 and Ibrutinib for Chronic Lymphocytic Leukemia. Hemasphere 2019; 3:e174. [PMID: 31723815 PMCID: PMC6746038 DOI: 10.1097/hs9.0000000000000174] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2018] [Revised: 12/19/2018] [Accepted: 12/28/2018] [Indexed: 12/21/2022] Open
|
426
|
Nguyen PH, Niesen E, Hallek M. New roles for B cell receptor associated kinases: when the B cell is not the target. Leukemia 2019; 33:576-587. [PMID: 30700840 DOI: 10.1038/s41375-018-0366-8] [Citation(s) in RCA: 30] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2018] [Accepted: 10/02/2018] [Indexed: 12/14/2022]
Abstract
Targeting of B cell receptor associated kinases (BAKs), such as Bruton's tyrosine kinase (BTK) or phosphoinositol-3-kinase (PI3K) delta, by specific inhibitors has revolutionized the therapy of B lymphoid malignancies. BAKs are critical signaling transducers of BCR signaling and seem relevant in B cell lymphoma pathogenesis. The functional relevance of BTK for lymphoid malignancies is strongly supported by the observation that resistance to therapy in CLL patients treated with BTK inhibitors such as ibrutinib is often associated with mutations in genes coding for BTK or Phospholipase-C gamma (PLCɣ). In some contrast, next generation sequencing data show that BAKs are mutated at very low frequency in treatment-naïve B cell lymphomas. Therefore, it remains debatable whether BAKs are essential drivers for lymphoma development. In addition, results obtained by targeted deletion of BAKs such as Lyn and Btk in murine CLL models suggest that BAKs may be essential to shape the dialogue between malignant B cells and the tumor microenvironment (TME). Since BAKs are expressed in multiple cell types, BAK inhibitors may disrupt the lymphoma supportive microenvironment. This concept also explains the typical response to BAK inhibitor treatment, characterized by a long-lasting increase of peripheral blood lymphoid cells, due to a redistribution from the lymphoid homing compartments. In addition, BAK inhibitors have shown some efficacy in solid tumors, probably through mediator cells in the TME. This review summarizes and validates the evidence for BAK inhibitors being part of a class of agents that modulate the (hematopoietic) microenvironment of cancers.
Collapse
Affiliation(s)
- Phuong-Hien Nguyen
- Department I of Internal Medicine, University Hospital of Cologne; Center for Integrated Oncology Cologne-Bonn; CECAD Center of Excellence on Cellular Stress Responses in Aging-Associated Diseases; Center for Molecular Medicine Cologne, University of Cologne, 50931, Cologne, Germany
| | - Emanuel Niesen
- Department I of Internal Medicine, University Hospital of Cologne; Center for Integrated Oncology Cologne-Bonn; CECAD Center of Excellence on Cellular Stress Responses in Aging-Associated Diseases; Center for Molecular Medicine Cologne, University of Cologne, 50931, Cologne, Germany
| | - Michael Hallek
- Department I of Internal Medicine, University Hospital of Cologne; Center for Integrated Oncology Cologne-Bonn; CECAD Center of Excellence on Cellular Stress Responses in Aging-Associated Diseases; Center for Molecular Medicine Cologne, University of Cologne, 50931, Cologne, Germany.
| |
Collapse
|
427
|
Le Bourgeois A. Gestion des toxicités des lymphocytes T exprimant un récepteur à l’antigène chimérique (CAR-T cells). Bull Cancer 2019; 105 Suppl 2:S188-S197. [PMID: 30686357 DOI: 10.1016/s0007-4551(19)30049-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/27/2022]
Abstract
TOXICITY MANAGEMENT OF CHIMERIC ANTIGEN RECEPTOR T-CELLS Chimeric antigen receptor T-cells (CAR T-cells) represent a very promising treatment for both hematological malignancies and solid tumors. Many clinical trials are currently on-going to establish the role of this emerging therapy. The toxicities observed after CAR T-cells infusion are mostly represented by a cytokine release syndrome and neurological symptoms. Athough reversible, these adverse events remain associated with morbidity and mortality in patients. The objective of this review is to describe these adverse events, also discussing their physiopathology and management, based on the recent recommendations published by the CARTOX working group (CAR T-cell-therapy-associated TOXicity working group) in the US. Cet article fait partie du numéro supplément Les cellules CAR-T : une révolution thérapeutique ? réalisé avec le soutien institutionnel des partenaires Gilead : Kite et Celgene.
Collapse
|
428
|
[CAR-T cells: Lymphocytes that express a chimeric antigen receptor]. Rev Med Interne 2019; 40:545-552. [PMID: 30686549 DOI: 10.1016/j.revmed.2018.12.002] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2018] [Revised: 11/30/2018] [Accepted: 12/04/2018] [Indexed: 12/27/2022]
Abstract
CAR-T cells are genetically modified human lymphocytes and gene therapy medicinal products. They are developed to treat cancers that express a membrane antigen targeted by the CAR. The FDA approved the two first-in-class medicinal products in 2017 and EMA in August 2018; both are autologous CAR-T cells targeting CD19 that is expressed at the surface of normal B-cells throughout their differentiation, and on B-cell lymphoid malignancies. Clinical efficacy was demonstrated for B-cell acute lymphoblastic leukemias, non-Hodgkin's lymphoma and chronic lymphocytic leukemia, although the marketing authorizations are less liberal in terms of indications. Manufacturing of these personalized treatments necessitates that a novel organization and supply chain be set in place, to ensure product preservation, patient safety and compliance with complex regulatory requirements. Side effects are commensurate with clinical efficacy and can be life-threatening: proper management imposes tight coordination between various specialists, particularly between hematologists and intensive care practitioners. High pricing for these treatments is part of a long-term trend for increasing costs of innovations in hematology and oncology; it questions the ability of healthcare systems to sustain their reimbursement.
Collapse
|
429
|
Kennedy VE, Muffly LS. Assessment of older adult candidates for allogeneic hematopoietic cell transplantation: updates and remaining questions. Expert Rev Hematol 2019; 12:99-106. [DOI: 10.1080/17474086.2019.1568236] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Affiliation(s)
- Vanessa E. Kennedy
- Department of Internal Medicine, Division of Hospital Medicine, Stanford University School of Medicine, Stanford, CA, USA
| | - Lori S. Muffly
- Department of Internal Medicine, Division of Blood and Marrow Transplantation, Stanford University School of Medicine, Stanford, CA, USA
| |
Collapse
|
430
|
Allan JN, Furman RR. Current trends in the management of Richter's syndrome. Int J Hematol Oncol 2019; 7:IJH09. [PMID: 30651968 PMCID: PMC6331753 DOI: 10.2217/ijh-2018-0010] [Citation(s) in RCA: 38] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2018] [Accepted: 11/21/2018] [Indexed: 12/16/2022] Open
Abstract
Richter's syndrome (RS) is a life-threatening complication of chronic lymphocytic leukemia (CLL). While previous research has increased our knowledge on the distinct evolutionary patterns of RS and provided a deeper understanding of the risk factors and molecular events predisposing to transformation, there remain few targetable aberrations and treatment is largely ineffective. The ability to obtain deeper remissions, without selecting for deletion 17p, by using novel B-cell receptor (BCR) antagonists and bcl2 inhibition might lead to a decrease in the incidence of RS, but these agents have done little to significantly change outcomes when incorporated into treatment regimens for RS. In this review we highlight the current landscape of molecular lesions specific to RS, review the data on historical treatment options, and look to the horizon for potential opportunities in the future.
Collapse
Affiliation(s)
- John N Allan
- Department of Medicine, Division of Hematology & Medical Oncology, New York-Presbyterian Hospital, Weill Cornell Medicine, 525 East 68th Street, New York, NY 10065, USA
| | - Richard R Furman
- Department of Medicine, Division of Hematology & Medical Oncology, New York-Presbyterian Hospital, Weill Cornell Medicine, 525 East 68th Street, New York, NY 10065, USA
| |
Collapse
|
431
|
Man F, Lim L, Volpe A, Gabizon A, Shmeeda H, Draper B, Parente-Pereira AC, Maher J, Blower PJ, Fruhwirth GO, T M de Rosales R. In Vivo PET Tracking of 89Zr-Labeled Vγ9Vδ2 T Cells to Mouse Xenograft Breast Tumors Activated with Liposomal Alendronate. Mol Ther 2019; 27:219-229. [PMID: 30429045 PMCID: PMC6318719 DOI: 10.1016/j.ymthe.2018.10.006] [Citation(s) in RCA: 79] [Impact Index Per Article: 13.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2018] [Revised: 10/03/2018] [Accepted: 10/08/2018] [Indexed: 12/19/2022] Open
Abstract
Gammadelta T (γδ-T) cells are strong candidates for adoptive immunotherapy in oncology due to their cytotoxicity, ease of expansion, and favorable safety profile. The development of γδ-T cell therapies would benefit from non-invasive cell-tracking methods and increased targeting to tumor sites. Here we report the use of [89Zr]Zr(oxinate)4 to track Vγ9Vδ2 T cells in vivo by positron emission tomography (PET). In vitro, we showed that 89Zr-labeled Vγ9Vδ2 T cells retained their viability, proliferative capacity, and anti-cancer cytotoxicity with minimal DNA damage for amounts of 89Zr ≤20 mBq/cell. Using a mouse xenograft model of human breast cancer, 89Zr-labeled γδ-T cells were tracked by PET imaging over 1 week. To increase tumor antigen expression, the mice were pre-treated with PEGylated liposomal alendronate. Liposomal alendronate, but not placebo liposomes or non-liposomal alendronate, significantly increased the 89Zr signal in the tumors, suggesting increased homing of γδ-T cells to the tumors. γδ-T cell trafficking to tumors occurred within 48 hr of administration. The presence of γδ-T cells in tumors, liver, and spleen was confirmed by histology. Our results demonstrate the suitability of [89Zr]Zr(oxinate)4 as a cell-labeling agent for therapeutic T cells and the potential benefits of liposomal bisphosphonate treatment before γδ-T cell administration.
Collapse
Affiliation(s)
- Francis Man
- School of Biomedical Engineering & Imaging Sciences, King's College London, St Thomas' Hospital, London SE1 7EH, UK
| | - Lindsay Lim
- School of Biomedical Engineering & Imaging Sciences, King's College London, St Thomas' Hospital, London SE1 7EH, UK
| | - Alessia Volpe
- School of Biomedical Engineering & Imaging Sciences, King's College London, St Thomas' Hospital, London SE1 7EH, UK
| | - Alberto Gabizon
- Oncology Institute, Shaare Zedek Medical Center and Hebrew University-School of Medicine, Jerusalem 9103102, Israel
| | - Hilary Shmeeda
- Oncology Institute, Shaare Zedek Medical Center and Hebrew University-School of Medicine, Jerusalem 9103102, Israel
| | - Benjamin Draper
- School of Cancer and Pharmaceutical Sciences, King's College London, Guy's Hospital, London SE1 9RT, UK
| | - Ana C Parente-Pereira
- School of Cancer and Pharmaceutical Sciences, King's College London, Guy's Hospital, London SE1 9RT, UK
| | - John Maher
- School of Cancer and Pharmaceutical Sciences, King's College London, Guy's Hospital, London SE1 9RT, UK
| | - Philip J Blower
- School of Biomedical Engineering & Imaging Sciences, King's College London, St Thomas' Hospital, London SE1 7EH, UK
| | - Gilbert O Fruhwirth
- School of Biomedical Engineering & Imaging Sciences, King's College London, St Thomas' Hospital, London SE1 7EH, UK
| | - Rafael T M de Rosales
- School of Biomedical Engineering & Imaging Sciences, King's College London, St Thomas' Hospital, London SE1 7EH, UK.
| |
Collapse
|
432
|
Mahadeo KM, Khazal SJ, Abdel-Azim H, Fitzgerald JC, Taraseviciute A, Bollard CM, Tewari P, Duncan C, Traube C, McCall D, Steiner ME, Cheifetz IM, Lehmann LE, Mejia R, Slopis JM, Bajwa R, Kebriaei P, Martin PL, Moffet J, McArthur J, Petropoulos D, O'Hanlon Curry J, Featherston S, Foglesong J, Shoberu B, Gulbis A, Mireles ME, Hafemeister L, Nguyen C, Kapoor N, Rezvani K, Neelapu SS, Shpall EJ. Management guidelines for paediatric patients receiving chimeric antigen receptor T cell therapy. Nat Rev Clin Oncol 2019; 16:45-63. [PMID: 30082906 PMCID: PMC7096894 DOI: 10.1038/s41571-018-0075-2] [Citation(s) in RCA: 155] [Impact Index Per Article: 25.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
In 2017, an autologous chimeric antigen receptor (CAR) T cell therapy indicated for children and young adults with relapsed and/or refractory CD19+ acute lymphoblastic leukaemia became the first gene therapy to be approved in the USA. This innovative form of cellular immunotherapy has been associated with remarkable response rates but is also associated with unique and often severe toxicities, which can lead to rapid cardiorespiratory and/or neurological deterioration. Multidisciplinary medical vigilance and the requisite health-care infrastructure are imperative to ensuring optimal patient outcomes, especially as these therapies transition from research protocols to standard care. Herein, authors representing the Pediatric Acute Lung Injury and Sepsis Investigators (PALISI) Network Hematopoietic Stem Cell Transplantation (HSCT) Subgroup and the MD Anderson Cancer Center CAR T Cell Therapy-Associated Toxicity (CARTOX) Program have collaborated to provide comprehensive consensus guidelines on the care of children receiving CAR T cell therapy.
Collapse
Affiliation(s)
- Kris M Mahadeo
- Department of Pediatrics, Stem Cell Transplantation and Cellular Therapy, CARTOX Program, The University of Texas MD Anderson Cancer Center, Houston, TX, USA.
| | - Sajad J Khazal
- Department of Pediatrics, Stem Cell Transplantation and Cellular Therapy, CARTOX Program, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Hisham Abdel-Azim
- Department of Pediatrics, Blood and Marrow Transplantation Program, Keck School of Medicine, University of Southern California, Children's Hospital Los Angeles, Los Angeles, CA, USA
| | - Julie C Fitzgerald
- Department of Anesthesiology and Critical Care, Division of Critical Care, University of Pennsylvania Perelman School of Medicine, Children's Hospital of Philadelphia, Philadelphia, PA, USA
| | - Agne Taraseviciute
- Department of Pediatrics, Division of Hematology-Oncology, University of Washington, Seattle Children's Hospital, Seattle, WA, USA
| | - Catherine M Bollard
- Center for Cancer and Immunology Research and Department of Pediatrics, Children's National and The George Washington University, Washington DC, USA
| | - Priti Tewari
- Department of Pediatrics, Stem Cell Transplantation, Baylor College of Medicine, Texas Children's Hospital, Houston, TX, USA
| | - Christine Duncan
- Pediatric Hematology-Oncology, Dana-Farber Cancer Institute, Harvard University, Boston, MA, USA
| | - Chani Traube
- Department of Pediatric Critical Care, Weil Cornell Medical College, New York Presbyterian Hospital, New York, NY, USA
| | - David McCall
- Department of Pediatrics, Stem Cell Transplantation and Cellular Therapy, CARTOX Program, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Marie E Steiner
- Department of Pediatrics, Division of Critical Care, University of Minnesota, Masonic Children's Hospital, University of Minnesota, Minneapolis, MN, USA
| | - Ira M Cheifetz
- Department of Pediatrics, Division of Critical Care, Duke Children's Hospital, Duke University, Durham, NC, USA
| | - Leslie E Lehmann
- Pediatric Hematology-Oncology, Dana-Farber Cancer Institute, Harvard University, Boston, MA, USA
| | - Rodrigo Mejia
- Department of Pediatrics, Critical Care, CARTOX Program, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - John M Slopis
- Department of Pediatrics, Neurology, CARTOX Program, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Rajinder Bajwa
- Department of Pediatrics, Division of Blood and Marrow Transplantation, Nationwide Children's Hospital, the Ohio State University, Columbus, OH, USA
| | - Partow Kebriaei
- Department of Stem Cell Transplantation and Cellular Therapy, CARTOX Program, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Paul L Martin
- Department of Pediatrics, Division of Blood and Marrow Transplant, Duke Children's Hospital, Duke University, Durham, NC, USA
| | - Jerelyn Moffet
- Department of Pediatrics, Division of Blood and Marrow Transplant, Duke Children's Hospital, Duke University, Durham, NC, USA
| | - Jennifer McArthur
- Department of Pediatrics, Division of Critical Care, St. Jude's Children's Research Hospital, Memphis, TN, USA
| | - Demetrios Petropoulos
- Department of Pediatrics, Stem Cell Transplantation and Cellular Therapy, CARTOX Program, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Joan O'Hanlon Curry
- Department of Pediatrics, Stem Cell Transplantation and Cellular Therapy, CARTOX Program, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Sarah Featherston
- Department of Pediatrics, Stem Cell Transplantation and Cellular Therapy, CARTOX Program, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Jessica Foglesong
- Department of Pediatrics, Stem Cell Transplantation and Cellular Therapy, CARTOX Program, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Basirat Shoberu
- Department of Pharmacy, Children's Hospital at Montefiore, Albert Einstein College of Medicine, Bronx, NY, USA
| | - Alison Gulbis
- Department of Pharmacy, CARTOX Program, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Maria E Mireles
- Department of Pharmacy, CARTOX Program, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Lisa Hafemeister
- Department of Pediatrics, Stem Cell Transplantation and Cellular Therapy, CARTOX Program, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Cathy Nguyen
- Department of Pediatrics, Stem Cell Transplantation and Cellular Therapy, CARTOX Program, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Neena Kapoor
- Department of Pediatrics, Blood and Marrow Transplantation Program, Keck School of Medicine, University of Southern California, Children's Hospital Los Angeles, Los Angeles, CA, USA
| | - Katayoun Rezvani
- Department of Stem Cell Transplantation and Cellular Therapy, CARTOX Program, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Sattva S Neelapu
- Department of Lymphoma and Myeloma, CARTOX Program, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Elizabeth J Shpall
- Department of Stem Cell Transplantation and Cellular Therapy, CARTOX Program, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| |
Collapse
|
433
|
Breman E, Demoulin B, Agaugué S, Mauën S, Michaux A, Springuel L, Houssa J, Huberty F, Jacques-Hespel C, Marchand C, Marijsse J, Nguyen T, Ramelot N, Violle B, Daro D, De Waele P, Gilham DE, Steenwinckel V. Overcoming Target Driven Fratricide for T Cell Therapy. Front Immunol 2018; 9:2940. [PMID: 30619300 PMCID: PMC6299907 DOI: 10.3389/fimmu.2018.02940] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2018] [Accepted: 11/30/2018] [Indexed: 12/14/2022] Open
Abstract
Chimeric Antigen Receptor (CAR) T cells expressing the fusion of the NKG2D protein with CD3ζ (NKG2D-CAR T Cells) acquire a specificity for stress-induced ligands expressed on hematological and solid cancers. However, these stress ligands are also transiently expressed by activated T cells implying that NKG2D-based T cells may undergo self-killing (fratricide) during cell manufacturing or during the freeze thaw cycle prior to infusion in patients. To avoid target-driven fratricide and enable the production of NKG2D-CAR T cells for clinical application, two distinct approaches were investigated. The first focused upon the inclusion of a Phosphoinositol-3-Kinase inhibitor (LY294002) into the production process. A second strategy involved the inclusion of antibody blockade of NKG2D itself. Both processes impacted T cell fratricide, albeit at different levels with the antibody process being the most effective in terms of cell yield. While both approaches generated comparable NKG2D-CAR T cells, there were subtle differences, for example in differentiation status, that were fine-tuned through the phasing of the inhibitor and antibody during culture in order to generate a highly potent NKG2D-CAR T cell product. By means of targeted inhibition of NKG2D expression or generic inhibition of enzyme function, target-driven CAR T fratricide can be overcome. These strategies have been incorporated into on-going clinical trials to enable a highly efficient and reproducible manufacturing process for NKG2D-CAR T cells.
Collapse
|
434
|
Abstract
Neurotoxicity is an important and common complication of chimeric antigen receptor-T cell therapies. Acute neurologic signs and/or symptoms occur in a significant proportion of patients treated with CD19-directed chimeric antigen receptor-T cells for B-cell malignancies. Clinical manifestations include headache, confusion, delirium, language disturbance, seizures and rarely, acute cerebral edema. Neurotoxicity is associated with cytokine release syndrome, which occurs in the setting of in-vivo chimeric antigen receptor-T cell activation and proliferation. The mechanisms that lead to neurotoxicity remain unknown, but data from patients and animal models suggest there is compromise of the blood-brain barrier, associated with high levels of cytokines in the blood and cerebrospinal fluid, as well as endothelial activation. Corticosteroids, interleukin-6-targeted therapies, and supportive care are frequently used to manage patients with neurotoxicity, but high-quality evidence of their efficacy is lacking.
Collapse
Affiliation(s)
- Juliane Gust
- Center for Integrative Brain Research, Seattle Children’s Research Institute, Seattle, WA, USA,Division of Pediatric Neurology, Department of Neurology, University of Washington, Seattle, WA, USA
| | - Agne Taraseviciute
- Department of Pediatrics, University of Southern California, Los Angeles, CA, USA
| | - Cameron J. Turtle
- Clinical Research Division and Integrated Immunotherapy Research Center, Fred Hutchinson Cancer Research Center, Seattle, WA, USA,Department of Medicine, University of Washington, Seattle, WA, USA
| |
Collapse
|
435
|
Appelbaum JS, Milano F. Hematopoietic Stem Cell Transplantation in the Era of Engineered Cell Therapy. Curr Hematol Malig Rep 2018; 13:484-493. [PMID: 30280289 PMCID: PMC6333424 DOI: 10.1007/s11899-018-0476-4] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
PURPOSE OF REVIEW Cellular therapy using T cells modified to express chimeric antigen receptors (CAR-T cells) has had striking success in patients that have failed previous treatment for CD19+ B cell non-Hodgkin lymphoma (NHL), chronic lymphocytic leukemia (CLL), or acute lymphoblastic leukemia (ALL). Curative therapy for this group of diseases has previously been limited to allogeneic hematopoietic cell transplantation HCT (alloHCT). The recent results of CAR-T cell therapy raise the question of how best to integrate CAR-T cell therapy and alloHCT in the care of these patients. RECENT FINDINGS Within the past 2 years, results from larger trials and increased follow-up of patients treated with CD19 CAR-T cell therapy suggest that some may achieve durable remission without transplant. The balance of efficacy and toxicity for CAR-T cell therapy and alloHCT vary by disease type, disease status at the time of treatment, patient characteristics, and the specific therapy employed. There are early signals that subsequent transplantation of patients who have achieved remission with CAR-T may be a potentially viable (though expensive) strategy.
Collapse
Affiliation(s)
- Jacob S Appelbaum
- Department of Medicine, University of Washington, 1100 Fairview AVE N, Mailstop # D5-100, Seattle, WA, 98109, USA.
- Fred Hutchinson Cancer Research Center, Seattle, WA, USA.
| | - Filippo Milano
- Department of Medicine, University of Washington, 1100 Fairview AVE N, Mailstop # D5-100, Seattle, WA, 98109, USA
- Fred Hutchinson Cancer Research Center, Seattle, WA, USA
| |
Collapse
|
436
|
Ding W. Richter transformation in the era of novel agents. HEMATOLOGY. AMERICAN SOCIETY OF HEMATOLOGY. EDUCATION PROGRAM 2018; 2018:256-263. [PMID: 30504319 PMCID: PMC6245983 DOI: 10.1182/asheducation-2018.1.256] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/13/2023]
Abstract
Recent approvals of several oral targeted agents have revolutionized chronic lymphocytic leukemia (CLL) therapy. However, CLL patients continue to progress; particularly, 4% to 20% of previously treated CLL patients undergo transformation into high-grade lymphoma. Richter transformation is defined as a transformation of CLL into aggressive lymphoma, most commonly diffuse large B-cell lymphoma. These patients typically have poor response to traditional chemotherapy used to treat de novo diffuse large B-cell lymphoma and similar or shorter overall survival (median 3-11 months) in the era of novel agents. Here, I review the contemporary literature on Richter transformation, particularly in the context of novel agents used in CLL, and discuss the management approach for these patients.
Collapse
MESH Headings
- Antineoplastic Agents/therapeutic use
- Disease-Free Survival
- Humans
- Leukemia, Lymphocytic, Chronic, B-Cell/drug therapy
- Leukemia, Lymphocytic, Chronic, B-Cell/metabolism
- Leukemia, Lymphocytic, Chronic, B-Cell/mortality
- Leukemia, Lymphocytic, Chronic, B-Cell/pathology
- Lymphoma, Large B-Cell, Diffuse/drug therapy
- Lymphoma, Large B-Cell, Diffuse/metabolism
- Lymphoma, Large B-Cell, Diffuse/mortality
- Lymphoma, Large B-Cell, Diffuse/pathology
- Survival Rate
Collapse
Affiliation(s)
- Wei Ding
- Chronic Lymphocytic Leukemia Group, Division of Hematology, Mayo Clinic, Rochester, MN
| |
Collapse
|
437
|
Chakraborty R, Sidana S, Shah GL, Scordo M, Hamilton BK, Majhail NS. Patient-Reported Outcomes with Chimeric Antigen Receptor T Cell Therapy: Challenges and Opportunities. Biol Blood Marrow Transplant 2018; 25:e155-e162. [PMID: 30500439 DOI: 10.1016/j.bbmt.2018.11.025] [Citation(s) in RCA: 58] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2018] [Accepted: 11/21/2018] [Indexed: 12/16/2022]
Abstract
Patient-reported outcomes (PROs) are an important tool to assess the impact of a new therapy on symptom burden and health-related quality of life (HRQoL). Chimeric antigen receptor T (CAR-T) cell therapies have been approved for use in relapsed or refractory leukemia and lymphoma based on promising efficacy in clinical trials. However, data are lacking on patient-reported toxicity and impact on HRQoL. This review provides an overview of the incorporation of PROs in CAR-T cell therapy and the specific challenges in this context. The first step is to demonstrate feasibility of PRO monitoring in the acute phase after CAR-T cell infusion. Apart from core PRO domains like physical functioning, disease-related symptoms, and symptomatic adverse effects, important measures to consider are cognitive functioning and financial toxicity. Because there are no validated PRO instruments in the setting of CAR-T cell therapy, universally validated measures like Patient-Reported Outcomes Measurement Information System (PROMIS) could be considered, which is also recommended in the setting of hematopoietic stem cell transplantation. Given the timeline of toxicities with CAR-T cell therapy, PRO instruments should be administered at baseline and at least weekly in the first 30 days. Subsequently, frequent monitoring of PROs in the first year might be helpful in identifying short- and intermediate-term toxicities, functional limitations, and neuropsychiatric effects. The major potential challenge in acute phase would be missing data when patients develop severe cytokine release syndrome or neurotoxicity. Designing a strategy for handling missing data is crucial. The long-term safety of CAR-T cell therapy is not well characterized because of short follow-up in most studies reported thus far. PROs should be measured at least yearly after the first year to identify potential late effects like cognitive deficit or autoimmune manifestations. Collaboration between institutions performing cellular therapy and engagement with patients, clinicians, and statisticians with expertise in PROs are crucial for setting a comprehensive agenda on integration of PROs with CAR-T cell therapy.
Collapse
Affiliation(s)
| | - Surbhi Sidana
- Division of Hematology, Mayo Clinic, Rochester, Minnesota
| | - Gunjan L Shah
- Adult Bone Marrow Transplant Service, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Michael Scordo
- Adult Bone Marrow Transplant Service, Memorial Sloan Kettering Cancer Center, New York, New York
| | | | | |
Collapse
|
438
|
Chimeric antigen receptor-modified T cell therapy in chronic lymphocytic leukemia. J Hematol Oncol 2018; 11:130. [PMID: 30458878 PMCID: PMC6247712 DOI: 10.1186/s13045-018-0676-3] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2018] [Accepted: 11/08/2018] [Indexed: 01/21/2023] Open
Abstract
Chronic lymphocytic leukemia (CLL), a common type of B cell chronic lymphoproliferative disorder in adults, has witnessed enormous development in its treatment in recent years. New drugs such as ibrutinib, idelalisib, and venetoclax have achieved great success in treating relapsed and refractory (R/R) CLL. In addition, with the development of immunotherapy, chimeric antigen receptor-engineered T cells (CAR-T) therapy, a novel adoptive immune treatment, has also become more and more important in treating R/R CLL. It combines the advantages of T cells and B cells via ex vivo gene transfer technology and is able to bind targets recognized by specific antibodies without antigen presentation, thus breaking the restriction of major histocompatibility complex. So far, there have been lots of studies exploring the application of CAR-T therapy in CLL. In this review, we describe the structure of chimeric antigen receptor, the preclinical, and clinical results of CAR-T therapy against CLL, along with its adverse events and advances in efficacy.
Collapse
|
439
|
Brudno JN, Kochenderfer JN. Recent advances in CAR T-cell toxicity: Mechanisms, manifestations and management. Blood Rev 2018; 34:45-55. [PMID: 30528964 DOI: 10.1016/j.blre.2018.11.002] [Citation(s) in RCA: 615] [Impact Index Per Article: 87.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2018] [Revised: 10/12/2018] [Accepted: 11/13/2018] [Indexed: 12/17/2022]
Abstract
Chimeric antigen receptor (CAR) T-cell therapy is an effective new treatment for hematologic malignancies. Two CAR T-cell products are now approved for clinical use by the U.S. FDA: tisagenlecleucel for pediatric acute lymphoblastic leukemia (ALL) and adult diffuse large B-cell lymphoma subtypes (DLBCL), and axicabtagene ciloleucel for DLBCL. CAR T-cell therapies are being developed for multiple myeloma, and clear evidence of clinical activity has been generated. A barrier to widespread use of CAR T-cell therapy is toxicity, primarily cytokine release syndrome (CRS) and neurologic toxicity. Manifestations of CRS include fevers, hypotension, hypoxia, end organ dysfunction, cytopenias, coagulopathy, and hemophagocytic lymphohistiocytosis. Neurologic toxicities are diverse and include encephalopathy, cognitive defects, dysphasias, seizures, and cerebral edema. Our understanding of the pathophysiology of CRS and neurotoxicity is continually improving. Early and peak levels of certain cytokines, peak blood CAR T-cell levels, patient disease burden, conditioning chemotherapy, CAR T-cell dose, endothelial activation, and CAR design are all factors that may influence toxicity. Multiple grading systems for CAR T-cell toxicity are in use; a universal grading system is needed so that CAR T-cell products can be compared across studies. Guidelines for toxicity management vary among centers, but typically include supportive care, plus immunosuppression with tocilizumab or corticosteroids administered for severe toxicity. Gaining a better understanding of CAR T-cell toxicities and developing new therapies for these toxicities are active areas of laboratory research. Further clinical investigation of CAR T-cell toxicity is also needed. In this review, we present guidelines for management of CRS and CAR neurotoxicity.
Collapse
Affiliation(s)
- Jennifer N Brudno
- Experimental Transplantation and Immunology Branch, National Cancer Institute, Building 10, Suite 3-3330, Bethesda, MD 20892, United States.
| | - James N Kochenderfer
- Experimental Transplantation and Immunology Branch, National Cancer Institute, Building 10, Suite 3-3330, Bethesda, MD 20892, United States.
| |
Collapse
|
440
|
Hay KA. Cytokine release syndrome and neurotoxicity after CD19 chimeric antigen receptor-modified (CAR-) T cell therapy. Br J Haematol 2018; 183:364-374. [PMID: 30407609 DOI: 10.1111/bjh.15644] [Citation(s) in RCA: 118] [Impact Index Per Article: 16.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/02/2023]
Abstract
Chimeric antigen receptor-modified (CAR)-T cells have demonstrated impressive results in the treatment of haematological malignancies. However, cytokine release syndrome (CRS) and neurotoxicity are common toxicities which are potentially life-threatening in severe cases. Risk factors for CRS and neurotoxicity identified so far include disease burden, lymphodepletion intensity and CAR-T cell dose administered. Risk-adapted dosing, with lower CAR-T cell doses administered to B-cell acute lymphoblastic leukaemia patients with high marrow blast counts, has been successful at decreasing severe CRS rates in this population. Intervention with therapies, such as tocilizumab and corticosteroids, have been effective at ameliorating toxicity, enabling CAR-T cells to be administered safely to many patients without significantly compromising efficacy. Deeper understanding of the pathophysiology of underlying CRS and neurotoxicity will enable the development of novel approaches to reduce toxicity and improve outcomes.
Collapse
Affiliation(s)
- Kevin A Hay
- Department of Medicine, University of British Columbia, Vancouver, BC, USA.,Fred Hutchinson Cancer Research Center, Seattle, WA, USA
| |
Collapse
|
441
|
Ti D, Niu Y, Wu Z, Fu X, Han W. Genetic engineering of T cells with chimeric antigen receptors for hematological malignancy immunotherapy. SCIENCE CHINA-LIFE SCIENCES 2018; 61:1320-1332. [DOI: 10.1007/s11427-018-9411-4] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/28/2018] [Accepted: 09/03/2018] [Indexed: 02/06/2023]
|
442
|
Hudecek M, Ivics Z. Non-viral therapeutic cell engineering with the Sleeping Beauty transposon system. Curr Opin Genet Dev 2018; 52:100-108. [PMID: 29957586 DOI: 10.1016/j.gde.2018.06.003] [Citation(s) in RCA: 44] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2018] [Revised: 04/30/2018] [Accepted: 06/04/2018] [Indexed: 12/28/2022]
Abstract
Widespread treatment of human diseases with gene therapies necessitates the development of gene transfer vectors that integrate genetic information effectively, safely and economically. Indeed, significant efforts have been devoted to engineer novel tools that (i) achieve high-level stable gene transfer at low toxicity to the host cell; (ii) induce low levels of genotoxicity and possess a `safe' integration profile with a high proportion of integrations into safe genomic locations; and (iii) are associated with acceptable cost per treatment, and scalable/exportable vector production to serve large numbers of patients. Two decades after the discovery of the Sleeping Beauty (SB) transposon, it has been transformed into a vector system that is fulfilling these requirements. Here we review recent developments in vectorization of SB as a tool for gene therapy, and highlight clinical development of the SB system towards hematopoietic stem cell gene therapy and cancer immunotherapy.
Collapse
Affiliation(s)
- Michael Hudecek
- Medizinische Klinik und Poliklinik II, Universitätsklinikum Würzburg, Würzburg, Germany
| | - Zoltán Ivics
- Division of Medical Biotechnology, Paul Ehrlich Institute, Langen, Germany.
| |
Collapse
|
443
|
Thomas X, Paubelle E. Tisagenlecleucel-T for the treatment of acute lymphocytic leukemia. Expert Opin Biol Ther 2018; 18:1095-1106. [DOI: 10.1080/14712598.2018.1533951] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Affiliation(s)
- Xavier Thomas
- Hospices Civils de Lyon, Hematology Department, Lyon-Sud Hospital, Pierre Bénite, France
| | - Etienne Paubelle
- Hospices Civils de Lyon, Hematology Department, Lyon-Sud Hospital, Pierre Bénite, France
| |
Collapse
|
444
|
Ramos JD, Yu EY. Immuno-oncology in urothelial carcinoma: who or what will ultimately sit on the iron throne? Immunotherapy 2018; 9:951-954. [PMID: 28971748 DOI: 10.2217/imt-2017-0109] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/02/2023] Open
Affiliation(s)
- Jorge D Ramos
- Fred Hutchinson Cancer Research Center, Seattle, WA, USA.,Department of Medicine, Division of Oncology, University of Washington School of Medicine, Seattle Cancer Care Alliance, 825 Eastlake Ave E - G4-800, Seattle, WA 98109-1024, USA
| | - Evan Y Yu
- Fred Hutchinson Cancer Research Center, Seattle, WA, USA.,Department of Medicine, Division of Oncology, University of Washington School of Medicine, Seattle Cancer Care Alliance, 825 Eastlake Ave E - G4-800, Seattle, WA 98109-1024, USA
| |
Collapse
|
445
|
Parikh SA. Chronic lymphocytic leukemia treatment algorithm 2018. Blood Cancer J 2018; 8:93. [PMID: 30283014 PMCID: PMC6170426 DOI: 10.1038/s41408-018-0131-2] [Citation(s) in RCA: 59] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2018] [Revised: 08/22/2018] [Accepted: 09/17/2018] [Indexed: 11/08/2022] Open
Abstract
The treatment landscape for patients with chronic lymphocytic leukemia (CLL) has changed considerably with the introduction of very effective oral targeted therapies (such as ibrutinib, idelalisib, and venetoclax), and next-generation anti-CD20 monoclonal antibodies (such as obinutuzumab). These agents lead to improved outcomes in CLL, even among patients with high-risk features, such as del17p13 or TP53 mutation and unmutated immunoglobulin heavy chain (IGHV) genes. Each of these treatments is associated with a unique toxicity profile; in the absence of randomized data, the choice of one type of treatment over another depends on the co-morbidities of the patient. Chemoimmunotherapy still plays an important role in the management of previously untreated CLL patients, particularly among young fit patients who have standard risk FISH profile and mutated IGHV genes. Richter's transformation of CLL remains a difficult complication to treat, although therapy with programmed death 1 inhibitors such as pembrolizumab and nivolumab has shown impressive responses in a subset of patients. Our ability to risk stratify CLL patients continues to evolve; the CLL-International Prognostic Index (CLL-IPI) is the best validated tool in predicting time to first therapy among previously untreated patients. This review summarizes the current approach to risk stratification and management of CLL patients.
Collapse
MESH Headings
- Algorithms
- Biomarkers, Tumor
- Combined Modality Therapy
- Disease Management
- Disease Progression
- Disease Susceptibility
- Drug Resistance, Neoplasm
- Humans
- Leukemia, Lymphocytic, Chronic, B-Cell/diagnosis
- Leukemia, Lymphocytic, Chronic, B-Cell/etiology
- Leukemia, Lymphocytic, Chronic, B-Cell/mortality
- Leukemia, Lymphocytic, Chronic, B-Cell/therapy
- Practice Guidelines as Topic
- Prognosis
- Recurrence
- Treatment Outcome
Collapse
|
446
|
Campbell JD, Fraser AR. Flow cytometric assays for identity, safety and potency of cellular therapies. CYTOMETRY PART B-CLINICAL CYTOMETRY 2018; 94:569-579. [DOI: 10.1002/cyto.b.21735] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/01/2017] [Revised: 06/18/2018] [Accepted: 07/10/2018] [Indexed: 12/12/2022]
|
447
|
Samples LS, Graf SA. On the front line: first choice pharmacotherapeutics for chronic lymphocytic leukemia. Expert Opin Pharmacother 2018; 19:1675-1684. [PMID: 30222470 DOI: 10.1080/14656566.2018.1524874] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/28/2022]
Abstract
INTRODUCTION Chronic lymphocytic leukemia (CLL) is a common hematologic malignancy with a highly variable clinical course. Frontline treatments include cytotoxic chemotherapies, immunotherapies, and small molecule inhibitors. Clinical and molecular factors guide treatment initiation and selection. Over the last decade, refinement of CLL risk stratification tools and growth of the arsenal of effective therapeutics have profoundly improved outcomes. These advances have concurrently increased the complexity of managing the early phases of treatment. AREAS COVERED This review describes the factors considered in the determination of first-line treatment of CLL. Areas of emphasis include assessment of patient fitness, disease classification and risk stratification, and the mechanisms, efficacy, and toxicities associated with available pharmacotherapeutics. EXPERT OPINION Multiple different treatments may be appropriate for a specific clinical scenario, and selection among them requires discussion of relative risks and benefits. Advances in frontline CLL treatment will continue to shift the treatment paradigm toward prioritizing quality of life alongside survival, limiting treatment and toxicity, and the development of biologically rational synergistic drug combinations and sequences.
Collapse
Affiliation(s)
- Laura S Samples
- a Department of Medicine , University of Washington School of Medicine , Seattle , WA , USA
| | - Solomon A Graf
- a Department of Medicine , University of Washington School of Medicine , Seattle , WA , USA.,b Clinical Research Division , Fred Hutchinson Cancer Research Center , Seattle , WA , USA.,c Department of Hematology and Oncology , Veterans Affairs Puget Sound Health Care System , Seattle , WA , USA
| |
Collapse
|
448
|
Improving CLL Vγ9Vδ2-T-cell fitness for cellular therapy by ex vivo activation and ibrutinib. Blood 2018; 132:2260-2272. [PMID: 30213872 DOI: 10.1182/blood-2017-12-822569] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2017] [Accepted: 08/01/2018] [Indexed: 12/27/2022] Open
Abstract
The efficacy of autologous (αβ) T-cell-based treatment strategies in chronic lymphocytic leukemia (CLL) has been modest. The Vγ9Vδ2-T cell subset consists of cytotoxic T lymphocytes with potent antilymphoma activity via a major histocompatibility complex-independent mechanism. We studied whether Vγ9Vδ2-T cells can be exploited as autologous effector lymphocytes in CLL. Healthy control Vγ9Vδ2-T cells were activated by and had potent cytolytic activity against CLL cells. However, CLL-derived Vγ9Vδ2-T cells proved dysfunctional with respect to effector cytokine production and degranulation, despite an increased frequency of the effector-type subset. Consequently, cytotoxicity against malignant B cells was hampered. A comparable dysfunctional phenotype was observed in healthy Vγ9Vδ2-T cells after coculture with CLL cells, indicating a leukemia-induced mechanism. Gene-expression profiling implicated alterations in synapse formation as a conceivable contributor to compromised Vγ9Vδ2-T-cell function in CLL patients. Dysfunction of Vγ9Vδ2-T cells was fully reversible upon activation with autologous monocyte-derived dendritic cells (moDCs). moDC activation resulted in efficient expansion and predominantly yielded Vγ9Vδ2-T cells with a memory phenotype. Furthermore, ibrutinib treatment promoted an antitumor T helper 1 (TH1) phenotype in Vγ9Vδ2-T cells, and we demonstrated binding of ibrutinib to IL-2-inducible kinase (ITK) in Vγ9Vδ2-T cells. Taken together, CLL-mediated dysfunction of autologous Vγ9Vδ2-T cells is fully reversible, resulting in potent cytotoxicity toward CLL cells. Our data support the potential use of Vγ9Vδ2-T cells as effector T cells in CLL immunotherapy and favor further exploration of combining Vγ9Vδ2-T-cell-based therapy with ibrutinib.
Collapse
|
449
|
Perez-Amill L, Marzal B, Urbano-Ispizua A, Juan M, Martín-Antonio B. CAR-T Cell Therapy: A Door Is Open to Find Innumerable Possibilities of Treatments for Cancer Patients. Turk J Haematol 2018; 35:217-228. [PMID: 30185400 PMCID: PMC6256819 DOI: 10.4274/tjh.2018.0196] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022] Open
Abstract
Seven years ago a chronic lymphocytic leukemia patient was for the first time successfully treated with chimeric antigen receptor (CAR)-modified T cells (CAR-T cells) to target CD19 overexpression in tumor cells. This was the beginning of the development of a new type of immunotherapy treatment in cancer patients. Since then, identification of novel antigens expressed in tumor cells and optimization of both CAR constructs and protocols of administration have opened up new avenues for the successful treatment of other hematological malignancies. However, research still continues to avoid some problems such as toxicities associated with the treatment and to find strategies to avoid tumor cell immune evasion mechanisms. On the other hand, for solid tumors, CAR-T therapy results are still in an early phase. In contrast to hematological malignancies, the complex tumor heterogeneity of solid tumors has led to the research of novel and challenging strategies to improve CAR-T cell activity. Here, we will review the main clinical results obtained with CAR-T cells in hematological malignancies, specifically focusing on CAR-T-19 and CAR-T against B-cell maturation antigen (CAR-T-BCMA). Moreover, we will mention the main problems that decrease CAR-T cell activity in solid tumors and the strategies to overcome them. Finally, we will present some of the first clinical results obtained for solid tumors.
Collapse
Affiliation(s)
- Lorena Perez-Amill
- Institut d’Investigacions Biomèdiques August Pi i Sunyer Hospital, Clinic of Hematology, Barcelona, Spain,Lorena Perez-Amill and Berta Marzal contributed to this article equally
| | - Berta Marzal
- Institut d’Investigacions Biomèdiques August Pi i Sunyer Hospital, Clinic of Immunology, Barcelona, Spain,Lorena Perez-Amill and Berta Marzal contributed to this article equally
| | - Alvaro Urbano-Ispizua
- Institut d’Investigacions Biomèdiques August Pi i Sunyer Hospital, Clinic of Hematology, Barcelona, Spain,Josep Carreras Leukaemia Research Institute, Barcelona, Spain
| | - Manel Juan
- Institut d’Investigacions Biomèdiques August Pi i Sunyer Hospital, Clinic of Immunology, Barcelona, Spain
| | - Beatriz Martín-Antonio
- Institut d’Investigacions Biomèdiques August Pi i Sunyer Hospital, Clinic of Hematology, Barcelona, Spain,Josep Carreras Leukaemia Research Institute, Barcelona, Spain
| |
Collapse
|
450
|
Met Ö, Jensen KM, Chamberlain CA, Donia M, Svane IM. Principles of adoptive T cell therapy in cancer. Semin Immunopathol 2018; 41:49-58. [PMID: 30187086 DOI: 10.1007/s00281-018-0703-z] [Citation(s) in RCA: 124] [Impact Index Per Article: 17.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2018] [Accepted: 08/13/2018] [Indexed: 01/01/2023]
Abstract
Adoptive cell therapy (ACT) utilizing either tumor-infiltrating lymphocyte (TIL)-derived T cells or T cells genetically engineered to express tumor recognizing receptors has emerged as a powerful and potentially curative therapy for several cancers. Many ACT-based therapies have recently entered late-phase clinical testing, with several T cell therapies already achieving regulatory approval for the treatment of patients with B cell malignancies. In this review, we briefly outline the principles of adoptively transferred T cells for the treatment of cancer.
Collapse
Affiliation(s)
- Özcan Met
- Center for Cancer Immune Therapy, Department of Hematology, Copenhagen University Hospital, Entrance 81, Floor 05, 2730, Herlev, Denmark.
- Department of Oncology, Copenhagen University Hospital, Herlev, Denmark.
- Department of Immunology and Microbiology, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark.
| | - Kasper Mølgaard Jensen
- Center for Cancer Immune Therapy, Department of Hematology, Copenhagen University Hospital, Entrance 81, Floor 05, 2730, Herlev, Denmark
| | - Christopher Aled Chamberlain
- Center for Cancer Immune Therapy, Department of Hematology, Copenhagen University Hospital, Entrance 81, Floor 05, 2730, Herlev, Denmark
| | - Marco Donia
- Center for Cancer Immune Therapy, Department of Hematology, Copenhagen University Hospital, Entrance 81, Floor 05, 2730, Herlev, Denmark
- Department of Oncology, Copenhagen University Hospital, Herlev, Denmark
| | - Inge Marie Svane
- Center for Cancer Immune Therapy, Department of Hematology, Copenhagen University Hospital, Entrance 81, Floor 05, 2730, Herlev, Denmark
- Department of Oncology, Copenhagen University Hospital, Herlev, Denmark
| |
Collapse
|