201
|
Mody H, Ogasawara K, Zhu X, Miles D, Shastri PN, Gokemeijer J, Liao MZ, Kasichayanula S, Yang TY, Chemuturi N, Gupta S, Jawa V, Upreti VV. Best Practices and Considerations for Clinical Pharmacology and Pharmacometric Aspects for Optimal Development of CAR-T and TCR-T Cell Therapies: An Industry Perspective. Clin Pharmacol Ther 2023; 114:530-557. [PMID: 37393588 DOI: 10.1002/cpt.2986] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2023] [Accepted: 06/26/2023] [Indexed: 07/04/2023]
Abstract
With the promise of a potentially "single dose curative" paradigm, CAR-T cell therapies have brought a paradigm shift in the treatment and management of hematological malignancies. Both CAR-T and TCR-T cell therapies have also made great progress toward the successful treatment of solid tumor indications. The field is rapidly evolving with recent advancements including the clinical development of "off-the-shelf" allogeneic CAR-T therapies that can overcome the long and difficult "vein-to-vein" wait time seen with autologous CAR-T therapies. There are unique clinical pharmacology, pharmacometric, bioanalytical, and immunogenicity considerations and challenges in the development of these CAR-T and TCR-T cell therapies. Hence, to help accelerate the development of these life-saving therapies for the patients with cancer, experts in this field came together under the umbrella of International Consortium for Innovation and Quality in Pharmaceutical Development (IQ) to form a joint working group between the Clinical Pharmacology Leadership Group (CPLG) and the Translational and ADME Sciences Leadership Group (TALG). In this white paper, we present the IQ consortium perspective on the best practices and considerations for clinical pharmacology and pharmacometric aspects toward the optimal development of CAR-T and TCR-T cell therapies.
Collapse
Affiliation(s)
- Hardik Mody
- Clinical Pharmacology, Genentech, South San Francisco, California, USA
| | - Ken Ogasawara
- Clinical Pharmacology, Pharmacometrics, Disposition and Bioanalysis, Bristol Myers Squibb, Lawrence Township, New Jersey, USA
| | - Xu Zhu
- Quantitative Clinical Pharmacology, AstraZeneca, Boston, Massachusetts, USA
| | - Dale Miles
- Clinical Pharmacology, Genentech, South San Francisco, California, USA
| | | | - Jochem Gokemeijer
- Discovery Biotherapeutics, Bristol Myers Squibb, Cambridge, Massachusetts, USA
| | - Michael Z Liao
- Clinical Pharmacology, Genentech, South San Francisco, California, USA
| | | | - Tong-Yuan Yang
- Bioanalytical Discovery and Development Sciences, Janssen R&D, LLC, Spring House, Pennsylvania, USA
| | - Nagendra Chemuturi
- Clinical Pharmacology, DMPK, Pharmacometrics, Moderna, Inc., Cambridge, Massachusetts, USA
| | - Swati Gupta
- Development Biological Sciences, Immunology, AbbVie, Irvine, California, USA
| | - Vibha Jawa
- Clinical Pharmacology, Pharmacometrics, Disposition and Bioanalysis, Bristol Myers Squibb, Lawrence Township, New Jersey, USA
| | - Vijay V Upreti
- Clinical Pharmacology, Modeling & Simulation, Amgen, South San Francisco, California, USA
| |
Collapse
|
202
|
Strati P, Gregory T, Majhail NS, Jain N. Chimeric Antigen Receptor T-Cell Therapy for Hematologic Malignancies: A Practical Review. JCO Oncol Pract 2023; 19:706-713. [PMID: 37406255 DOI: 10.1200/op.22.00819] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2022] [Revised: 03/09/2023] [Accepted: 05/24/2023] [Indexed: 07/07/2023] Open
Abstract
Chimeric antigen receptor T-cell (CAR-T) therapy has become an established therapeutic approach for the treatment of hematologic malignancies. The field continues to evolve rapidly and newer-generation constructs are being designed to enhance proliferative capacity, and achieve long-term persistence and greater efficacy with an overall lower incidence of toxicity. Initial clinical application of CAR-T therapies has focused on relapsed and/or refractory hematologic malignancies, and Food and Drug Administration-approved CAR-T products targeting CD19 are available for B-cell acute lymphoblastic leukemia and low- and high-grade B-cell non-Hodgkin lymphoma, and targeting B-cell maturation antigen are available for multiple myeloma. Cytokine release syndrome and immune effector cell-associated neurotoxicity syndrome have been recognized as class specific toxicities associated with these novel therapies. In this review, we focus on the clinical application of CAR-T therapies in adult patients with hematologic malignancies, including access issues, outpatient administration, and appropriate timing for referring a patient to a CAR-T treatment center.
Collapse
Affiliation(s)
- Paolo Strati
- Department of Lymphoma and Myeloma, The University of Texas MD Anderson Cancer Center, Houston, TX
| | - Tara Gregory
- Colorado Blood Cancer Institute, Denver, CO
- Sarah Cannon Transplant and Cellular Therapy Program at Presbyterian/St Luke's Medical Center, Denver, CO
| | - Navneet S Majhail
- Sarah Cannon, Nashville, TN
- Sarah Cannon Transplant and Cellular Therapy Program at TriStar Centennial, Nashville, TN
| | - Nitin Jain
- Department of Leukemia, The University of Texas MD Anderson Cancer Center, Houston, TX
| |
Collapse
|
203
|
Thiruvengadam SK, Shouse G, Danilov AV. Thinking "outside the germinal center": Re-educating T cells to combat follicular lymphoma. Blood Rev 2023; 61:101099. [PMID: 37173225 DOI: 10.1016/j.blre.2023.101099] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2023] [Revised: 04/04/2023] [Accepted: 05/08/2023] [Indexed: 05/15/2023]
Abstract
There have been significant advancements in the management of follicular lymphoma (FL), the most common indolent lymphoma. These include immunomodulatory agents such as lenalidomide, epigenetic modifiers (tazemetostat), and phosphoinotiside-3 kinase inhibitors (copanlisib). The focus of this review is T cell-engager therapies, namely chimeric antigen receptor (CAR) T-cell therapy and bispecific antibodies, have recently transformed the treatment landscape of FL. Two CAR T cell products, axicabtagene ciloleucel (axi-cel) and tisagenlecleucel (tisa-cel), and one bispecific antibody, mosunetuzumab, recently received FDA approvals in FL. Several other new immune effector drugs are being evaluated and will expand the treatment armamentarium. This review focuses on CAR T-cell and bispecific antibody therapies, details their safety and efficacy and considers their evolving role in the current treatment landscape of FL.
Collapse
|
204
|
Abstract
PURPOSE OF REVIEW The purpose of this review is to present updates in the field of bispecific antibodies focusing on those agents that have been recently approved for multiple myeloma, follicular lymphoma and diffuse large B cell lymphoma. RECENT FINDINGS Teclistamab, the β-cell maturation antigen -targeted bispecific antibody has shown efficacy and tolerability in the fourth line setting for multiple myeloma. Mosunetuzumab, the CD20-targeted bispecific antibody has shown excellent response rates and durability in third line and beyond follicular lymphoma. Epcoritamab and glofitamab have both shown excellent response rates in heavily pretreated patients with diffuse large B cell lymphoma including those with prior chimeric antigen receptor T cell therapy. The toxicity is significant but manageable for both agents. Epcoritamab is approved by the FDA in the United States, while glofitamab is approved for use in Canada for patients with diffuse large B cell lymphoma refractory to 2 or more prior lines of therapy. SUMMARY Bispecific antibodies represent a novel therapeutic resource that is poised to dramatically change the treatment landscape of many hematologic malignancies, but so far, initial successes include multiple myeloma, follicular lymphoma, and diffuse large B cell lymphoma, where several agents have been recently approved.
Collapse
Affiliation(s)
- Geoffrey Shouse
- City of Hope Comprehensive Cancer Center, Duarte, California, USA
| |
Collapse
|
205
|
Springell D, O'Reilly M, Roddie C. Supportive care for chimeric antigen receptor T-cell patients. Curr Opin Support Palliat Care 2023; 17:231-239. [PMID: 37418578 DOI: 10.1097/spc.0000000000000657] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/09/2023]
Abstract
PURPOSE OF REVIEW The purpose of this review is to provide clear guidance to health professionals delivering chimeric antigen receptor T-cell (CAR-T) therapy on the best supportive management throughout the CAR-T pathway, from referral to long-term follow-up, including psychosocial aspects. RECENT FINDINGS CAR-T therapy has changed the treatment landscape for relapsed/refractory (r/r) B-cell malignancy. Approximately 40% of r/r B-cell leukaemia/lymphoma patients receiving CD19-targeted CAR-T therapy achieve durable remission following a single dose. The field is rapidly expanding to encompass new CAR-T products for indications such as multiple myeloma, mantle cell lymphoma and follicular lymphoma, and the number of patients eligible to receive CAR-T therapy is likely to continue to grow exponentially. CAR-T therapy is logistically challenging to deliver, with involvement of many stakeholders. In many cases, CAR-T therapy requires an extended inpatient hospital admission, particularly in older, comorbid patients, and is associated with potentially severe immune side effects. Further, CAR-T therapy can lead to protracted cytopenias that can last for several months accompanied by a susceptibility to infection. SUMMARY For the reasons listed above, standardised, comprehensive supportive care is critically important to ensure that CAR-T therapy is delivered as safely as possible and that patients are fully informed of the risks and benefits, as well as the requirement for extended hospital admission and follow-up, to fully realise the potential of this transformative treatment modality.
Collapse
Affiliation(s)
| | - Maeve O'Reilly
- Department of Haematology, University College London Hospital
| | - Claire Roddie
- Department of Haematology, University College London Hospital
- Research Department of Haematology, University College London, London, UK
| |
Collapse
|
206
|
Gabrielli G, Shouval R, Ghilardi G, van den Brink M, Ruella M. Harnessing the Gut Microbiota to Potentiate the Efficacy of CAR T Cell Therapy. Hemasphere 2023; 7:e950. [PMID: 37637993 PMCID: PMC10448936 DOI: 10.1097/hs9.0000000000000950] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2023] [Accepted: 07/27/2023] [Indexed: 08/29/2023] Open
Affiliation(s)
- Giulia Gabrielli
- Center for Cellular Immunotherapies, University of Pennsylvania School of Medicine, Philadelphia, PA, USA
- Lymphoma Program, Abramson Cancer Center, University of Pennsylvania, Philadelphia, PA, USA
- Department of Molecular Biotechnology and Health Sciences, University of Torino, Italy
- Division of Hematology-Oncology, Department of Medicine, University of Pennsylvania School of Medicine, Philadelphia, PA, USA
| | - Roni Shouval
- Adult Bone Marrow Transplant Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA
- Department of Medicine, Weill Cornell Medical College, New York, NY, USA
| | - Guido Ghilardi
- Center for Cellular Immunotherapies, University of Pennsylvania School of Medicine, Philadelphia, PA, USA
- Lymphoma Program, Abramson Cancer Center, University of Pennsylvania, Philadelphia, PA, USA
- Division of Hematology-Oncology, Department of Medicine, University of Pennsylvania School of Medicine, Philadelphia, PA, USA
| | - Marcel van den Brink
- Adult Bone Marrow Transplant Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA
- Department of Medicine, Weill Cornell Medical College, New York, NY, USA
| | - Marco Ruella
- Center for Cellular Immunotherapies, University of Pennsylvania School of Medicine, Philadelphia, PA, USA
- Lymphoma Program, Abramson Cancer Center, University of Pennsylvania, Philadelphia, PA, USA
- Division of Hematology-Oncology, Department of Medicine, University of Pennsylvania School of Medicine, Philadelphia, PA, USA
| |
Collapse
|
207
|
Rajamäki A, Hujo M, Sund R, Prusila RE, Kuusisto ME, Kuitunen H, Jantunen E, Mercadal S, Sorigue M, Sancho JM, Sunela K, Kuittinen O. Estimating the Lifetime Treatment Burden of Patients With Follicular Lymphoma: A Retrospective Study Using Real-World Multicenter Data. JCO Clin Cancer Inform 2023; 7:e2300067. [PMID: 37756639 PMCID: PMC10569776 DOI: 10.1200/cci.23.00067] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2023] [Revised: 06/16/2023] [Accepted: 07/05/2023] [Indexed: 09/29/2023] Open
Abstract
PURPOSE Although follicular lymphoma is characterized by long natural history and frequent relapses, data on the number of patients receiving subsequent therapy lines are scarce. To perform reliable health economical calculations for various treatment options, data regarding the lifetime number of therapy courses are needed. The purpose of this study was to use real-world data to create a model that could estimate the treatment burden over a 20-year period. MATERIALS AND METHODS We performed a 20-year simulation on the basis of retrospectively obtained multicenter data of 743 patients with follicular lymphoma. The simulation was carried out in two steps: First, a competing risk model on the basis of Weibull distribution was used to simulate the state transitions from diagnosis onward and from first-line therapy onward. Then, the data were completed by imputing on the basis of the existing data. Completion of data was repeated for 1,000 times to estimate reliability. RESULTS In 20 years, 97% (2.5-97.5 percentile range: 96%-98%), 66% (61%-70%), 34% (30%-41%), and 15% (9%-18%) of the patients received first-line, second-line, third-line, and fourth-line therapies, respectively. The median number of therapy lines received by each patient was two. CONCLUSION Despite long remissions, approximately two thirds of the patients receive at least two lines and one-third at least three lines of therapy during their lifetime.
Collapse
Affiliation(s)
- Aino Rajamäki
- Institute of Clinical Medicine, Faculty of Health Sciences, University of Eastern Finland, Kuopio, Finland
| | - Mika Hujo
- School of Computing, Faculty of Science and Forestry, University of Eastern Finland, Kuopio, Finland
| | - Reijo Sund
- Institute of Clinical Medicine, Faculty of Health Sciences, University of Eastern Finland, Kuopio, Finland
| | - Roosa E.I. Prusila
- Medical Research Center and Cancer and Translational Research Unit, University of Oulu and Oulu University Hospital, Oulu, Finland
| | - Milla E.L. Kuusisto
- Medical Research Center and Cancer and Translational Research Unit, University of Oulu and Oulu University Hospital, Oulu, Finland
| | - Hanne Kuitunen
- Department of Oncology, Oulu University Hospital, Oulu, Finland
| | - Esa Jantunen
- Institute of Clinical Medicine, Faculty of Health Sciences, University of Eastern Finland, Kuopio, Finland
- Department of Medicine, Kuopio University Hospital, Kuopio, Finland
| | | | - Marc Sorigue
- Medical Department, Trialing Health, Barcelona, Spain
| | - Juan-Manuel Sancho
- Department of Hematology, ICO-Hospital Germans Trias i Pujol, IJC, UAB, Badalona, Spain
| | - Kaisa Sunela
- Finnish Medicines Agency Fimea, Tampere, Finland
| | - Outi Kuittinen
- Institute of Clinical Medicine, Faculty of Health Sciences, University of Eastern Finland, Kuopio, Finland
- Medical Research Center and Cancer and Translational Research Unit, University of Oulu and Oulu University Hospital, Oulu, Finland
- Department of Oncology, Kuopio University Hospital, Kuopio, Finland
| |
Collapse
|
208
|
Tang L, Huang Z, Mei H, Hu Y. Immunotherapy in hematologic malignancies: achievements, challenges and future prospects. Signal Transduct Target Ther 2023; 8:306. [PMID: 37591844 PMCID: PMC10435569 DOI: 10.1038/s41392-023-01521-5] [Citation(s) in RCA: 23] [Impact Index Per Article: 23.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2022] [Revised: 05/31/2023] [Accepted: 06/04/2023] [Indexed: 08/19/2023] Open
Abstract
The immune-cell origin of hematologic malignancies provides a unique avenue for the understanding of both the mechanisms of immune responsiveness and immune escape, which has accelerated the progress of immunotherapy. Several categories of immunotherapies have been developed and are being further evaluated in clinical trials for the treatment of blood cancers, including stem cell transplantation, immune checkpoint inhibitors, antigen-targeted antibodies, antibody-drug conjugates, tumor vaccines, and adoptive cell therapies. These immunotherapies have shown the potential to induce long-term remission in refractory or relapsed patients and have led to a paradigm shift in cancer treatment with great clinical success. Different immunotherapeutic approaches have their advantages but also shortcomings that need to be addressed. To provide clinicians with timely information on these revolutionary therapeutic approaches, the comprehensive review provides historical perspectives on the applications and clinical considerations of the immunotherapy. Here, we first outline the recent advances that have been made in the understanding of the various categories of immunotherapies in the treatment of hematologic malignancies. We further discuss the specific mechanisms of action, summarize the clinical trials and outcomes of immunotherapies in hematologic malignancies, as well as the adverse effects and toxicity management and then provide novel insights into challenges and future directions.
Collapse
Affiliation(s)
- Lu Tang
- Institute of Hematology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, 430022, Wuhan, China
- Hubei Clinical Medical Center of Cell Therapy for Neoplastic Disease, 430022, Wuhan, China
- Key Laboratory of Biological Targeted Therapy, the Ministry of Education, 430022, Wuhan, China
| | - Zhongpei Huang
- Institute of Hematology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, 430022, Wuhan, China
- Hubei Clinical Medical Center of Cell Therapy for Neoplastic Disease, 430022, Wuhan, China
- Key Laboratory of Biological Targeted Therapy, the Ministry of Education, 430022, Wuhan, China
| | - Heng Mei
- Institute of Hematology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, 430022, Wuhan, China.
- Hubei Clinical Medical Center of Cell Therapy for Neoplastic Disease, 430022, Wuhan, China.
- Key Laboratory of Biological Targeted Therapy, the Ministry of Education, 430022, Wuhan, China.
- Hubei Key Laboratory of Biological Targeted Therapy, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, 430022, Wuhan, China.
| | - Yu Hu
- Institute of Hematology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, 430022, Wuhan, China.
- Hubei Clinical Medical Center of Cell Therapy for Neoplastic Disease, 430022, Wuhan, China.
- Key Laboratory of Biological Targeted Therapy, the Ministry of Education, 430022, Wuhan, China.
- Hubei Key Laboratory of Biological Targeted Therapy, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, 430022, Wuhan, China.
| |
Collapse
|
209
|
Detroit M, Collier M, Beeker N, Willems L, Decroocq J, Deau-Fischer B, Vignon M, Birsen R, Moufle F, Leclaire C, Balladur E, Deschamps P, Chauchet A, Batista R, Limat S, Treluyer JM, Ricard L, Stocker N, Hermine O, Choquet S, Morel V, Metz C, Bouscary D, Kroemer M, Zerbit J. Predictive Factors of Response to Immunotherapy in Lymphomas: A Multicentre Clinical Data Warehouse Study (PRONOSTIM). Cancers (Basel) 2023; 15:4028. [PMID: 37627056 PMCID: PMC10452259 DOI: 10.3390/cancers15164028] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2023] [Revised: 08/04/2023] [Accepted: 08/07/2023] [Indexed: 08/27/2023] Open
Abstract
Immunotherapy (IT) is a major therapeutic strategy for lymphoma, significantly improving patient prognosis. IT remains ineffective for a significant number of patients, however, and exposes them to specific toxicities. The identification predictive factors around efficacy and toxicity would allow better targeting of patients with a higher ratio of benefit to risk. PRONOSTIM is a multicenter and retrospective study using the Clinical Data Warehouse (CDW) of the Greater Paris University Hospitals network. Adult patients with Hodgkin lymphoma or diffuse large-cell B lymphoma treated with immune checkpoint inhibitors or CAR T (Chimeric antigen receptor T) cells between 2017 and 2022 were included. Analysis of covariates influencing progression-free survival (PFS) or the occurrence of grade ≥3 toxicity was performed. In total, 249 patients were included. From this study, already known predictors for response or toxicity of CAR T cells such as age, elevated lactate dehydrogenase, and elevated C-Reactive Protein at the time of infusion were confirmed. In addition, male gender, low hemoglobin, and hypo- or hyperkalemia were demonstrated to be potential predictive factors for progression after CAR T cell therapy. These findings prove the attractiveness of CDW in generating real-world data, and show its essential contribution to identifying new predictors for decision support before starting IT.
Collapse
Affiliation(s)
- Marion Detroit
- Pharmacy Department, Pitié-Salpêtrière Hospital, Greater Paris University Hospitals (AP-HP), Sorbonne University, 75013 Paris, France; (M.D.); (C.M.)
| | - Mathis Collier
- Clinical Research Unit, Cochin Hospital, AP-HP, Centre Paris-Cité University, 75014 Paris, France; (M.C.); (N.B.); (J.-M.T.)
| | - Nathanaël Beeker
- Clinical Research Unit, Cochin Hospital, AP-HP, Centre Paris-Cité University, 75014 Paris, France; (M.C.); (N.B.); (J.-M.T.)
| | - Lise Willems
- Hematology Department, Cochin Hospital, AP-HP, Centre Paris-Cité University, 75014 Paris, France; (L.W.); (J.D.); (B.D.-F.); (M.V.); (R.B.); (D.B.)
| | - Justine Decroocq
- Hematology Department, Cochin Hospital, AP-HP, Centre Paris-Cité University, 75014 Paris, France; (L.W.); (J.D.); (B.D.-F.); (M.V.); (R.B.); (D.B.)
| | - Bénédicte Deau-Fischer
- Hematology Department, Cochin Hospital, AP-HP, Centre Paris-Cité University, 75014 Paris, France; (L.W.); (J.D.); (B.D.-F.); (M.V.); (R.B.); (D.B.)
| | - Marguerite Vignon
- Hematology Department, Cochin Hospital, AP-HP, Centre Paris-Cité University, 75014 Paris, France; (L.W.); (J.D.); (B.D.-F.); (M.V.); (R.B.); (D.B.)
| | - Rudy Birsen
- Hematology Department, Cochin Hospital, AP-HP, Centre Paris-Cité University, 75014 Paris, France; (L.W.); (J.D.); (B.D.-F.); (M.V.); (R.B.); (D.B.)
| | - Frederique Moufle
- Adult Department, Hospital at Home, AP-HP, Centre Paris-Cité University, 75014 Paris, France; (F.M.); (C.L.); (E.B.)
| | - Clément Leclaire
- Adult Department, Hospital at Home, AP-HP, Centre Paris-Cité University, 75014 Paris, France; (F.M.); (C.L.); (E.B.)
| | - Elisabeth Balladur
- Adult Department, Hospital at Home, AP-HP, Centre Paris-Cité University, 75014 Paris, France; (F.M.); (C.L.); (E.B.)
| | - Paul Deschamps
- Hematology Oncology Department, André Mignot Hospital, 78157 Le Chesnay, France;
| | - Adrien Chauchet
- Hematology Department, University Hospital of Besançon, 25000 Besançon, France;
| | - Rui Batista
- Pharmacy Department, Cochin Hospital, AP-HP, Centre Paris-Cité University, 75014 Paris, France;
| | - Samuel Limat
- Pharmacy Department, University Hospital of Besançon, 25000 Besançon, France; (S.L.); (M.K.)
- French National Institute of Health and Medical Research (INSERM), Etablissement Français du Sang Bourgogne Franche-Comte (EFS BFC), UMR1098, RIGHT, University of Bourgogne Franche-Comté, 25000 Besançon, France
| | - Jean-Marc Treluyer
- Clinical Research Unit, Cochin Hospital, AP-HP, Centre Paris-Cité University, 75014 Paris, France; (M.C.); (N.B.); (J.-M.T.)
- Regional Pharmacovigilance Center, Pharmacology Department, Cochin Hospital, AP-HP, Centre Paris-Cité University, 75014 Paris, France
| | - Laure Ricard
- Hematology Department, Saint Antoine Hospital, AP-HP, INSERM UMRs 938, Sorbonne University, 75012 Paris, France; (L.R.); (N.S.)
| | - Nicolas Stocker
- Hematology Department, Saint Antoine Hospital, AP-HP, INSERM UMRs 938, Sorbonne University, 75012 Paris, France; (L.R.); (N.S.)
| | - Olivier Hermine
- Hematology Department, Necker Hospital, AP-HP, Centre Paris-Cité University, 75015 Paris, France;
| | - Sylvain Choquet
- Hematology Department, Pitié-Salpêtrière Hospital, AP-HP, Sorbonne University, 75013 Paris, France; (S.C.); (V.M.)
| | - Véronique Morel
- Hematology Department, Pitié-Salpêtrière Hospital, AP-HP, Sorbonne University, 75013 Paris, France; (S.C.); (V.M.)
| | - Carole Metz
- Pharmacy Department, Pitié-Salpêtrière Hospital, Greater Paris University Hospitals (AP-HP), Sorbonne University, 75013 Paris, France; (M.D.); (C.M.)
| | - Didier Bouscary
- Hematology Department, Cochin Hospital, AP-HP, Centre Paris-Cité University, 75014 Paris, France; (L.W.); (J.D.); (B.D.-F.); (M.V.); (R.B.); (D.B.)
| | - Marie Kroemer
- Pharmacy Department, University Hospital of Besançon, 25000 Besançon, France; (S.L.); (M.K.)
- French National Institute of Health and Medical Research (INSERM), Etablissement Français du Sang Bourgogne Franche-Comte (EFS BFC), UMR1098, RIGHT, University of Bourgogne Franche-Comté, 25000 Besançon, France
| | - Jérémie Zerbit
- Cancer Treatment Unit, Pharmacy Department, Hospital at Home, AP-HP, Centre Paris-Cité University, 75014 Paris, France
| |
Collapse
|
210
|
Bücklein V, Perez A, Rejeski K, Iacoboni G, Jurinovic V, Holtick U, Penack O, Kharboutli S, Blumenberg V, Ackermann J, Frölich L, Johnson G, Patel K, Arciola B, Mhaskar R, Wood A, Schmidt C, Albanyan O, Gödel P, Hoster E, Bullinger L, Mackensen A, Locke F, von Bergwelt M, Barba P, Subklewe M, Jain MD. Inferior Outcomes of EU Versus US Patients Treated With CD19 CAR-T for Relapsed/Refractory Large B-cell Lymphoma: Association With Differences in Tumor Burden, Systemic Inflammation, Bridging Therapy Utilization, and CAR-T Product Use. Hemasphere 2023; 7:e907. [PMID: 37449196 PMCID: PMC10337711 DOI: 10.1097/hs9.0000000000000907] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2022] [Accepted: 05/03/2023] [Indexed: 07/18/2023] Open
Abstract
Real-world evidence suggests a trend toward inferior survival of patients receiving CD19 chimeric antigen receptor (CAR) T-cell therapy in Europe (EU) and with tisagenlecleucel. The underlying logistic, patient- and disease-related reasons for these discrepancies remain poorly understood. In this multicenter retrospective observational study, we studied the patient-individual journey from CAR-T indication to infusion, baseline features, and survival outcomes in 374 patients treated with tisagenlecleucel (tisa-cel) or axicabtagene-ciloleucel (axi-cel) in EU and the United States (US). Compared with US patients, EU patients had prolonged indication-to-infusion intervals (66 versus 50 d; P < 0.001) and more commonly received intermediary therapies (holding and/or bridging therapy, 94% in EU versus 74% in US; P < 0.001). Baseline lactate dehydrogenase (LDH) (median 321 versus 271 U/L; P = 0.02) and ferritin levels (675 versus 425 ng/mL; P = 0.004) were significantly elevated in the EU cohort. Overall, we observed inferior survival in EU patients (median progression-free survival [PFS] 3.1 versus 9.2 months in US; P < 0.001) and with tisa-cel (3.2 versus 9.2 months with axi-cel; P < 0.001). On multivariate Lasso modeling, nonresponse to bridging, elevated ferritin, and increased C-reactive protein represented independent risks for treatment failure. Weighing these variables into a patient-individual risk balancer (high risk [HR] balancer), we found higher levels in EU versus US and tisa-cel versus axi-cel cohorts. Notably, superior PFS with axi-cel was exclusively evident in patients at low risk for progression (according to the HR balancer), but not in high-risk patients. These data demonstrate that inferior survival outcomes in EU patients are associated with longer time-to-infusion intervals, higher tumor burden/LDH levels, increased systemic inflammatory markers, and CAR-T product use.
Collapse
Affiliation(s)
- Veit Bücklein
- Department of Medicine III, University Hospital, LMU Munich, Germany
- Laboratory for Translational Cancer Immunology, LMU Gene Center, Munich, Germany
| | - Ariel Perez
- Department of Blood and Marrow Transplant and Cellular Immunotherapy, Moffitt Cancer Center, Tampa, FL, USA
- Blood and Marrow Transplant Program, Miami Cancer Institute, Miami, FL, USA
| | - Kai Rejeski
- Department of Medicine III, University Hospital, LMU Munich, Germany
- Laboratory for Translational Cancer Immunology, LMU Gene Center, Munich, Germany
- German Cancer Consortium (DKTK) Munich Site, and German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Gloria Iacoboni
- Department of Hematology, Vall d’Hebron Institute of Oncology (VHIO), University Hospital Vall d’Hebron, Department of Medicine, Universitat Autònoma of Barcelona (UAB), Spain
| | - Vindi Jurinovic
- Institute for Medical Information Processing, Biometry, and Epidemiology, LMU Munich, Germany
| | - Udo Holtick
- Department I of Internal Medicine, Medical Faculty and University Hospital, Cologne, University of Cologne, Germany
| | - Olaf Penack
- Charité – Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Department of Hematology, Oncology and Tumorimmunology, Berlin, Germany
| | - Soraya Kharboutli
- Department of Internal Medicine 5, Hematology and Oncology, University of Erlangen-Nuremberg, Germany
| | - Viktoria Blumenberg
- Department of Medicine III, University Hospital, LMU Munich, Germany
- Laboratory for Translational Cancer Immunology, LMU Gene Center, Munich, Germany
- German Cancer Consortium (DKTK) Munich Site, and German Cancer Research Center (DKFZ), Heidelberg, Germany
| | | | - Lisa Frölich
- Department of Medicine III, University Hospital, LMU Munich, Germany
| | - Grace Johnson
- USF Health Morsani College of Medicine, Tampa, FL, USA
| | - Kedar Patel
- USF Health Morsani College of Medicine, Tampa, FL, USA
| | - Brian Arciola
- USF Health Morsani College of Medicine, Tampa, FL, USA
| | - Rahul Mhaskar
- USF Health Morsani College of Medicine, Tampa, FL, USA
| | - Anthony Wood
- Department of Blood and Marrow Transplant and Cellular Immunotherapy, Moffitt Cancer Center, Tampa, FL, USA
| | - Christian Schmidt
- Department of Medicine III, University Hospital, LMU Munich, Germany
| | - Omar Albanyan
- Department of Blood and Marrow Transplant and Cellular Immunotherapy, Moffitt Cancer Center, Tampa, FL, USA
| | - Philipp Gödel
- Department I of Internal Medicine, Medical Faculty and University Hospital, Cologne, University of Cologne, Germany
| | - Eva Hoster
- Institute for Medical Information Processing, Biometry, and Epidemiology, LMU Munich, Germany
| | - Lars Bullinger
- Charité – Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Department of Hematology, Oncology and Tumorimmunology, Berlin, Germany
| | - Andreas Mackensen
- Department of Internal Medicine 5, Hematology and Oncology, University of Erlangen-Nuremberg, Germany
| | - Frederick Locke
- Department of Blood and Marrow Transplant and Cellular Immunotherapy, Moffitt Cancer Center, Tampa, FL, USA
| | - Michael von Bergwelt
- Department of Medicine III, University Hospital, LMU Munich, Germany
- German Cancer Consortium (DKTK) Munich Site, and German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Pere Barba
- Department of Hematology, Vall d’Hebron Institute of Oncology (VHIO), University Hospital Vall d’Hebron, Department of Medicine, Universitat Autònoma of Barcelona (UAB), Spain
| | - Marion Subklewe
- Department of Medicine III, University Hospital, LMU Munich, Germany
- Laboratory for Translational Cancer Immunology, LMU Gene Center, Munich, Germany
- German Cancer Consortium (DKTK) Munich Site, and German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Michael D. Jain
- Department of Blood and Marrow Transplant and Cellular Immunotherapy, Moffitt Cancer Center, Tampa, FL, USA
| |
Collapse
|
211
|
Rafaniello C, Liguori V, Zinzi A, Gaio M, Falco A, Di Costanzo L, Gargano F, Trimarco V, Cataldi M, Capuano A. A Pharmacovigilance Study on the Safety of Axicabtagene Ciloleucel Based on Spontaneous Reports from the EudraVigilance Database. Biomedicines 2023; 11:2162. [PMID: 37626659 PMCID: PMC10452324 DOI: 10.3390/biomedicines11082162] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2023] [Revised: 07/27/2023] [Accepted: 07/29/2023] [Indexed: 08/27/2023] Open
Abstract
During pre-approval clinical trials, the safety of axi-cel, a second-generation CAR-T-cell therapy directed against CD19, which dramatically improved the prognosis of intractable B-cell lymphomas, has been investigated only in about 400 patients. Therefore, additional information on this issue is urgently needed. In the present paper, we evaluated the 2905 ICSRs with axi-cel as the suspected drug that had been uploaded in the EudraVigilance database from 1 January 2018 to 31 December 2022. About 80% of the reported adverse events were serious, and about 20% of them did not fully resolve or caused death. The adverse events most-frequently reported were Nervous system disorders (25.6%) and, among them, immune-effector-cell-associated neurotoxicity syndrome, followed by Immune system disorders (23.1%), General disorders and administration site conditions (12.0%), Blood and lymphatic system disorders (7.2%), and Infections and infestations (5.8%). Disproportionality analysis showed that the frequency of reported adverse events related to the nervous system was higher with axi-cel than with the other approved CAR-T-cells, except brexu-cel. In conclusion, real-world pharmacovigilance data showed that nervous system and immune system disorders are the adverse events most reported in axi-cel-related ICSRs and suggest that axi-cel could be more neurotoxic than other CAR-T-cells.
Collapse
Affiliation(s)
- Concetta Rafaniello
- Campania Regional Centre for Pharmacovigilance and Pharmacoepidemiology, 80138 Naples, Italy; (C.R.); (V.L.); (A.Z.); (M.G.); (A.F.)
- Section of Pharmacology “L. Donatelli”, Department of Experimental Medicine, University of Campania “Luigi Vanvitelli”, 80138 Naples, Italy;
| | - Valerio Liguori
- Campania Regional Centre for Pharmacovigilance and Pharmacoepidemiology, 80138 Naples, Italy; (C.R.); (V.L.); (A.Z.); (M.G.); (A.F.)
- Section of Pharmacology “L. Donatelli”, Department of Experimental Medicine, University of Campania “Luigi Vanvitelli”, 80138 Naples, Italy;
| | - Alessia Zinzi
- Campania Regional Centre for Pharmacovigilance and Pharmacoepidemiology, 80138 Naples, Italy; (C.R.); (V.L.); (A.Z.); (M.G.); (A.F.)
- Section of Pharmacology “L. Donatelli”, Department of Experimental Medicine, University of Campania “Luigi Vanvitelli”, 80138 Naples, Italy;
| | - Mario Gaio
- Campania Regional Centre for Pharmacovigilance and Pharmacoepidemiology, 80138 Naples, Italy; (C.R.); (V.L.); (A.Z.); (M.G.); (A.F.)
- Section of Pharmacology “L. Donatelli”, Department of Experimental Medicine, University of Campania “Luigi Vanvitelli”, 80138 Naples, Italy;
| | - Angela Falco
- Campania Regional Centre for Pharmacovigilance and Pharmacoepidemiology, 80138 Naples, Italy; (C.R.); (V.L.); (A.Z.); (M.G.); (A.F.)
- Section of Pharmacology, Department of Neuroscience, Reproductive Sciences and Dentistry, Federico II University of Naples, Via Sergio Pansini 5, 80131 Naples, Italy;
| | - Luigi Di Costanzo
- Section of Pharmacology “L. Donatelli”, Department of Experimental Medicine, University of Campania “Luigi Vanvitelli”, 80138 Naples, Italy;
| | - Francesca Gargano
- Department of Anesthesia and Resuscitation, Biomedical Campus University of Rome, 00128 Rome, Italy;
| | - Valentina Trimarco
- Section of Pharmacology, Department of Neuroscience, Reproductive Sciences and Dentistry, Federico II University of Naples, Via Sergio Pansini 5, 80131 Naples, Italy;
| | - Mauro Cataldi
- Section of Pharmacology, Department of Neuroscience, Reproductive Sciences and Dentistry, Federico II University of Naples, Via Sergio Pansini 5, 80131 Naples, Italy;
| | - Annalisa Capuano
- Campania Regional Centre for Pharmacovigilance and Pharmacoepidemiology, 80138 Naples, Italy; (C.R.); (V.L.); (A.Z.); (M.G.); (A.F.)
- Section of Pharmacology “L. Donatelli”, Department of Experimental Medicine, University of Campania “Luigi Vanvitelli”, 80138 Naples, Italy;
| |
Collapse
|
212
|
Johnson PC, Dhawale T, Newcomb RA, Amonoo HL, Lavoie MW, Vaughn D, Karpinski K, El-Jawahri A. Longitudinal patient-reported outcomes in patients receiving chimeric antigen receptor T-cell therapy. Blood Adv 2023; 7:3541-3550. [PMID: 36995091 PMCID: PMC10368828 DOI: 10.1182/bloodadvances.2022009117] [Citation(s) in RCA: 10] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2022] [Revised: 01/25/2023] [Accepted: 02/13/2023] [Indexed: 03/31/2023] Open
Abstract
Chimeric antigen receptor T-cell therapy (CAR-T) has transformed the treatment for relapsed/refractory hematologic malignancies; however, data on patient-reported outcomes in CAR-T are limited. We conducted a longitudinal study of adults with hematologic malignancies receiving CAR-T. We assessed quality of life (QOL; functional assessment of cancer therapy-general), psychological distress (hospital anxiety and depression scale, patient health questionnaire-9, posttraumatic stress disorder [PTSD] checklist), and physical symptoms (Edmonton symptom assessment scale-revised) at baseline, 1 week, 1, 3, and 6 months after CAR-T. We used linear mixed models to identify factors associated with QOL trajectory. We enrolled 103 of 142 eligible patients (3 did not receive CAR-T). QOL (B = 1.96; P < .001) and depression (B = -0.32; P = .001) worsened by 1 week and improved by 6 months after CAR-T. At 6 months, 18%, 22%, and 22% reported clinically significant depression, anxiety, and PTSD symptoms, respectively. At 1 week, 52% reported severe physical symptoms, declining to 28% at 6 months after CAR-T. In unadjusted linear mixed models, worse Eastern Cooperative Oncology Group performance status (B = 1.24; P = .042), receipt of tocilizumab (B = 1.54; P = .042), and receipt of corticosteroids for cytokine release syndrome and/or neurotoxicity (B = 2.05; P = .006) were associated with higher QOL trajectory. After CAR-T, QOL declined, and depression increased early, followed by improvements in QOL, psychological distress, and physical symptoms by 6 months after infusion. A significant minority of patients reported substantial psychological distress and physical symptoms longitudinally.
Collapse
Affiliation(s)
- P. Connor Johnson
- Division of Hematology & Oncology, Department of Medicine, Massachusetts General Hospital, Boston, MA
- Harvard Medical School, Boston, MA
| | - Tejaswini Dhawale
- Division of Hematology & Oncology, Department of Medicine, Massachusetts General Hospital, Boston, MA
- Harvard Medical School, Boston, MA
| | - Richard A. Newcomb
- Division of Hematology & Oncology, Department of Medicine, Massachusetts General Hospital, Boston, MA
- Harvard Medical School, Boston, MA
| | - Hermioni L. Amonoo
- Harvard Medical School, Boston, MA
- Department of Psychiatry, Brigham and Women’s Hospital, Boston, MA
- Department of Psychosocial Oncology and Palliative Care, Dana-Farber Cancer Institute, Boston, MA
| | - Mitchell W. Lavoie
- University of Massachusetts Chan Medical School, Worchester, MA
- Department of Psychiatry, Massachusetts General Hospital, Boston, MA
| | - Dagny Vaughn
- Department of Psychiatry, Massachusetts General Hospital, Boston, MA
- University of Tennessee Health Science Center College of Medicine, Memphis, TN
| | - Kyle Karpinski
- Department of Psychiatry, Massachusetts General Hospital, Boston, MA
| | - Areej El-Jawahri
- Division of Hematology & Oncology, Department of Medicine, Massachusetts General Hospital, Boston, MA
- Harvard Medical School, Boston, MA
| |
Collapse
|
213
|
[Chinese guidelines for diagnosis and treatment of follicular lymphoma(2023)]. ZHONGHUA XUE YE XUE ZA ZHI = ZHONGHUA XUEYEXUE ZAZHI 2023; 44:529-534. [PMID: 37749030 PMCID: PMC10509619 DOI: 10.3760/cma.j.issn.0253-2727.2023.07.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Subscribe] [Scholar Register] [Received: 05/16/2023] [Indexed: 09/27/2023]
|
214
|
Zheng Z, Li S, Liu M, Chen C, Zhang L, Zhou D. Fine-Tuning through Generations: Advances in Structure and Production of CAR-T Therapy. Cancers (Basel) 2023; 15:3476. [PMID: 37444586 DOI: 10.3390/cancers15133476] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2023] [Revised: 06/23/2023] [Accepted: 06/27/2023] [Indexed: 07/15/2023] Open
Abstract
Chimeric antigen receptor (CAR)-T cell therapy is a promising form of immunotherapy that has seen significant advancements in the past few decades. It involves genetically modifying T cells to target cancer cells expressing specific antigens, providing a novel approach to treating various types of cancer. However, the initial success of first-generation CAR-T cells was limited due to inadequate proliferation and undesirable outcomes. Nonetheless, significant progress has been made in CAR-T cell engineering, leading to the development of the latest fifth-generation CAR-T cells that can target multiple antigens and overcome individual limitations. Despite these advancements, some shortcomings prevent the widespread use of CAR-T therapy, including life-threatening toxicities, T-cell exhaustion, and inadequate infiltration for solid tumors. Researchers have made considerable efforts to address these issues by developing new strategies for improving CAR-T cell function and reducing toxicities. This review provides an overview of the path of CAR-T cell development and highlights some of the prominent advances in its structure and manufacturing process, which include the strategies to improve antigen recognition, enhance T-cell activation and persistence, and overcome immune escape. Finally, the review briefly covers other immune cells for cancer therapy and ends with the discussion on the broad prospects of CAR-T in the treatment of various diseases, not just hematological tumors, and the challenges that need to be addressed for the widespread clinical application of CAR-T cell therapies.
Collapse
Affiliation(s)
- Zhibo Zheng
- Department of International Medical Services, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100730, China
| | - Siyuan Li
- Department of Hematology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100730, China
| | - Mohan Liu
- Department of Breast Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100730, China
| | - Chuyan Chen
- Department of Gastroenterology, National Clinical Research Center for Digestive Disease, Beijing Digestive Disease Center, Beijing Friendship Hospital, Capital Medical University, Beijing 100730, China
| | - Lu Zhang
- Department of Hematology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100730, China
| | - Daobin Zhou
- Department of Hematology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100730, China
| |
Collapse
|
215
|
Sauter CS, Hill BT. Rush Hour Update: Roadblocks to CARs on the Expressway. Transplant Cell Ther 2023; 29:408-409. [PMID: 37400191 DOI: 10.1016/j.jtct.2023.06.005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/05/2023]
Affiliation(s)
- Craig S Sauter
- Department of Hematology and Medical Oncology, Taussig Cancer Institute, Cleveland Clinic, Cleveland, Ohio
| | - Brian T Hill
- Department of Hematology and Medical Oncology, Taussig Cancer Institute, Cleveland Clinic, Cleveland, Ohio
| |
Collapse
|
216
|
Rejeski K, Jain MD, Smith EL. Mechanisms of Resistance and Treatment of Relapse after CAR T-cell Therapy for Large B-cell Lymphoma and Multiple Myeloma. Transplant Cell Ther 2023; 29:418-428. [PMID: 37076102 PMCID: PMC10330792 DOI: 10.1016/j.jtct.2023.04.007] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2023] [Revised: 04/04/2023] [Accepted: 04/11/2023] [Indexed: 04/21/2023]
Abstract
Although chimeric antigen receptor (CAR) T cell therapy (CAR-T) has altered the treatment landscape for relapsed/refractory B cell malignancies and multiple myeloma, only a minority of patients attain long-term disease remission. The underlying reasons for CAR-T resistance are multifaceted and can be broadly divided into host-related, tumor-intrinsic, microenvironmental and macroenvironmental, and CAR-T-related factors. Emerging host-related determinants of response to CAR-T relate to gut microbiome composition, intact hematopoietic function, body composition, and physical reserve. Emerging tumor-intrinsic resistance mechanisms include complex genomic alterations and mutations to immunomodulatory genes. Furthermore, the extent of systemic inflammation prior to CAR-T is a potent biomarker of response and reflects a proinflammatory tumor micromilieu characterized by infiltration of myeloid-derived suppressor cells and regulatory T cell populations. The tumor and its surrounding micromilieu also can shape the response of the host to CAR-T infusion and the subsequent expansion and persistence of CAR T cells, a prerequisite for efficient eradication of tumor cells. Here, focusing on both large B cell lymphoma and multiple myeloma, we review resistance mechanisms, explore therapeutic avenues to overcome resistance to CAR-T, and discuss the management of patients who relapse after CAR-T.
Collapse
Affiliation(s)
- Kai Rejeski
- Department of Medicine III – Hematology/Oncology, University Hospital, LMU Munich, Munich, Germany
- German Cancer Consortium (DKTK), Munich Site, and German Cancer Research Center, Heidelberg, Germany
| | - Michael D. Jain
- Department of Blood and Marrow Transplant and Cellular Immunotherapy, Moffitt Cancer Center, Tampa, USA
| | | |
Collapse
|
217
|
Gazeau N, Liang EC, Wu QV, Voutsinas JM, Barba P, Iacoboni G, Kwon M, Ortega JLR, López-Corral L, Hernani R, Ortiz-Maldonado V, Martínez-Cibrian N, Martinez AP, Maziarz RT, Williamson S, Nemecek ER, Shadman M, Cowan AJ, Green DJ, Kimble E, Hirayama AV, Maloney DG, Turtle CJ, Gauthier J. Anakinra for Refractory Cytokine Release Syndrome or Immune Effector Cell-Associated Neurotoxicity Syndrome after Chimeric Antigen Receptor T Cell Therapy. Transplant Cell Ther 2023; 29:430-437. [PMID: 37031746 PMCID: PMC10330552 DOI: 10.1016/j.jtct.2023.04.001] [Citation(s) in RCA: 12] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2023] [Revised: 04/02/2023] [Accepted: 04/02/2023] [Indexed: 04/11/2023]
Abstract
Chimeric antigen receptor-engineered (CAR)-T cell therapy remains limited by significant toxicities, including cytokine release syndrome (CRS) and immune effector cell-associated neurotoxicity syndrome (ICANS). The optimal management of severe and/or refractory CRS/ICANS remains ill-defined. Anakinra has emerged as a promising agent based on preclinical data, but its safety and efficacy in CAR-T therapy recipients are unknown. The primary objective of this study was to evaluate the safety of anakinra to treat refractory CRS and ICANS after CAR-T therapy. The secondary objective was to evaluate the impact of key treatment-, patient-, and disease-related variables on the time to CRS/ICANS resolution and treatment-related mortality (TRM). We retrospectively analyzed the outcomes of 43 patients with B cell or plasma cell malignancies treated with anakinra for refractory CRS or ICANS at 9 institutions in the United States and Spain between 2019 and 2022. Cause-specific Cox regression was used to account for competing risks. Multivariable cause-specific Cox regression was used to estimate the effect of anakinra dose on outcomes while minimizing treatment allocation bias by including age, CAR-T product, prelymphodepletion (pre-LD) ferritin, and performance status. Indications for anakinra treatment were grade ≥2 ICANS with worsening or lack of symptom improvement despite treatment with high-dose corticosteroids (n = 40) and grade ≥2 CRS with worsening symptoms despite treatment with tocilizumab (n = 3). Anakinra treatment was feasible and safe; discontinuation of therapy because of anakinra-related side effects was reported in only 3 patients (7%). The overall response rate (ORR) to CAR-T therapy was 77%. The cumulative incidence of TRM in the whole cohort was 7% (95% confidence interval [CI], 2% to 17%) at 28 days and 23% (95% CI, 11% to 38%) at 60 days after CAR-T infusion. The cumulative incidence of TRM at day 28 after initiation of anakinra therapy was 0% in the high-dose (>200 mg/day i.v.) recipient group and 47% (95% CI, 20% to 70%) in the low-dose (100 to 200 mg/day s.c. or i.v.) recipient group. The median cumulative incidence of CRS/ICANS resolution from the time of anakinra initiation was 7 days in the high-dose group and was not reached in the low-dose group, owing to the high TRM in this group. Univariate Cox modeling suggested a shorter time to CRS/ICANS resolution in the high-dose recipients (hazard ratio [HR], 2.19; 95% CI, .94 to 5.12; P = .069). In a multivariable Cox model for TRM including age, CAR-T product, pre-LD ferritin level, and pre-LD Karnofsky Performance Status (KPS), higher anakinra dose remained associated with lower TRM (HR, .41 per 1 mg/kg/day increase; 95% CI, .17 to .96; P = .039. The sole factor independently associated with time to CRS/ICANS resolution in a multivariable Cox model including age, CAR-T product, pre-LD ferritin and anakinra dose was higher pre-LD KPS (HR, 1.05 per 10% increase; 95% CI, 1.01 to 1.09; P = .02). Anakinra treatment for refractory CRS or ICANS was safe at doses up to 12 mg/kg/day i.v. We observed an ORR of 77% after CAR-T therapy despite anakinra treatment, suggesting a limited impact of anakinra on CAR-T efficacy. Higher anakinra dose may be associated with faster CRS/ICANS resolution and was independently associated with lower TRM. Prospective comparative studies are needed to confirm our findings.
Collapse
Affiliation(s)
- Nicolas Gazeau
- Clinical Research Division, Fred Hutchinson Cancer Center, Seattle, Washington; Hematology Service, Centre Hospitalier Universitaire de Lille, Lille, France
| | - Emily C Liang
- Clinical Research Division, Fred Hutchinson Cancer Center, Seattle, Washington; Department of Medicine, University of Washington, Seattle, Washington
| | - Qian Vicky Wu
- Clinical Research Division, Fred Hutchinson Cancer Center, Seattle, Washington
| | - Jenna M Voutsinas
- Clinical Research Division, Fred Hutchinson Cancer Center, Seattle, Washington
| | - Pere Barba
- Hematology Department, Hospital Universitari Vall d'Hebron. Universitat Autonoma de Barcelona, Barcelona, Spain
| | - Gloria Iacoboni
- Hematology Department, Hospital Universitari Vall d'Hebron. Universitat Autonoma de Barcelona, Barcelona, Spain
| | - Mi Kwon
- Department of Hematology, Hospital General Universitario Gregrorio Marañon, Institute of Health Research Gregorio Marañon. Universidad Complutense de Madrid, Madrid, Spain
| | - Juan Luis Reguera Ortega
- Hematology Service, Hospital Universitario Virgen del Rocío, Instituto de Biomedicina de Sevilla, Sevilla, Spain
| | - Lucía López-Corral
- Hematology Department, Complejo Asistencial Universitario de Salamanca-IBSAL, Centro de Investigación del Cáncer-IBMCC, Spain
| | - Rafael Hernani
- Hematology Department, Hospital Clinico Universitario, INCLIVA Research Institut, Valencia, Spain
| | | | | | | | - Richard T Maziarz
- Center for Hematologic Malignancies, Knight Cancer Institute, Oregon Health and Science University, Portland, Oregon
| | - Staci Williamson
- Center for Hematologic Malignancies, Knight Cancer Institute, Oregon Health and Science University, Portland, Oregon
| | - Eneida R Nemecek
- Center for Hematologic Malignancies, Knight Cancer Institute, Oregon Health and Science University, Portland, Oregon
| | - Mazyar Shadman
- Clinical Research Division, Fred Hutchinson Cancer Center, Seattle, Washington; Department of Medicine, University of Washington, Seattle, Washington
| | - Andrew J Cowan
- Clinical Research Division, Fred Hutchinson Cancer Center, Seattle, Washington; Department of Medicine, University of Washington, Seattle, Washington
| | - Damian J Green
- Clinical Research Division, Fred Hutchinson Cancer Center, Seattle, Washington; Department of Medicine, University of Washington, Seattle, Washington
| | - Erik Kimble
- Clinical Research Division, Fred Hutchinson Cancer Center, Seattle, Washington; Department of Medicine, University of Washington, Seattle, Washington
| | - Alexandre V Hirayama
- Clinical Research Division, Fred Hutchinson Cancer Center, Seattle, Washington; Department of Medicine, University of Washington, Seattle, Washington
| | - David G Maloney
- Clinical Research Division, Fred Hutchinson Cancer Center, Seattle, Washington; Department of Medicine, University of Washington, Seattle, Washington
| | - Cameron J Turtle
- Clinical Research Division, Fred Hutchinson Cancer Center, Seattle, Washington; Department of Medicine, University of Washington, Seattle, Washington; Sydney Medical School, Faculty of Medicine and Health, The University of Sydney, Camperdown, New South Wales, Australia
| | - Jordan Gauthier
- Clinical Research Division, Fred Hutchinson Cancer Center, Seattle, Washington; Department of Medicine, University of Washington, Seattle, Washington.
| |
Collapse
|
218
|
Ahmed N, Wesson W, Mushtaq MU, Porter DL, Nasta SD, Brower J, Bachanova V, Hu M, Nastoupil LJ, Oluwole OO, Patel VG, Oliai C, Riedell PA, Bishop MR, Shah GL, Perales MA, Schachter L, Maziarz RT, McGuirk JP. Patient Characteristics and Outcomes of Outpatient Tisagenlecleucel Recipients for B Cell Non-Hodgkin Lymphoma. Transplant Cell Ther 2023; 29:449.e1-449.e7. [PMID: 37120134 PMCID: PMC11027185 DOI: 10.1016/j.jtct.2023.04.019] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2023] [Revised: 04/15/2023] [Accepted: 04/24/2023] [Indexed: 05/01/2023]
Abstract
Tisagenlecleucel (tisa-cel) is an approved CD19-directed chimeric antigen receptor T cell (CAR-T) therapy for relapsed/refractory B cell malignancies. Given potentially life-threatening toxicities, including cytokine release syndrome and immune effector cell-associated neurotoxicity syndrome, inpatient tisa-cel infusion and toxicity monitoring are often considered; however, the toxicity profile of tisa-cel may be conducive to outpatient administration. Here we review the characteristics and outcomes of tisa-cel recipients treated in the outpatient setting. Patients age ≥18 years with B cell non-Hodgkin lymphoma who received tisa-cel between June 25, 2018, and January 22, 2021, at 9 US academic medical centers were included in a retrospective analysis. Six of the 9 representative centers (75%) had an outpatient program in place. A total of 157 patients were evaluable, including 93 (57%) in the outpatient treatment group and 64 (43%) in the inpatient treatment group. Baseline characteristics, toxicity and efficacy, and resource utilization were summarized. The most common lymphodepletion (LD) regimen was bendamustine in the outpatient group (65%) and fludarabine/cyclophosphamide (91%) in the inpatient group. The outpatient group had more patients with a Charlson Comorbidity Index of 0 (51% versus 15%; P < .001), fewer patients with an elevated lactate dehydrogenase (LDH) level above the normal range at the time of LD (32% versus 57%, P = .003) compared to the inpatient group, and a lower Endothelial Activation and Stress Index score (.57 versus 1.4; P < .001). Any-grade CRS and ICANS were lower in the outpatient group (29% versus 56% [P < .001] and 10% versus 16% [P = .051], respectively). Forty-two outpatient tisa-cel recipients (45%) required an unplanned admission, with a median length of stay of 5 days (range, 1 to 27 days), compared to 13 days (range, 4 to 38 days) in the inpatient group. The median number of tocilizumab doses administered was similar in the 2 groups as were the rate of intensive care unit (ICU) transfer (5% versus 8%; P = .5) and median length of ICU stay (6 days versus 5 days; P = .7). There were no toxicity-related deaths in the 30 days post-CAR-T infusion in either group. Progression-free survival and overall survival were similar in the 2 groups. With careful patient selection, outpatient tisa-cel administration is feasible and associated with similar efficacy outcomes as inpatient treatment. Outpatient toxicity monitoring and management may help optimize healthcare resource utilization.
Collapse
Affiliation(s)
- Nausheen Ahmed
- University of Kansas Medical Center, Kansas City, Kansas.
| | - William Wesson
- University of Kansas Medical Center, Kansas City, Kansas
| | | | - David L Porter
- University of Pennsylvania and Abramson Cancer Center, Philadelphia, Pennsylvania
| | - Sunita D Nasta
- University of Pennsylvania and Abramson Cancer Center, Philadelphia, Pennsylvania
| | - Jamie Brower
- University of Pennsylvania and Abramson Cancer Center, Philadelphia, Pennsylvania
| | - Veronika Bachanova
- Division of Hematology, Oncology and Transplantation, Department of Medicine, University of Minnesota, Minneapolis, Minnesota
| | - Marie Hu
- Division of Hematology, Oncology and Transplantation, Department of Medicine, University of Minnesota, Minneapolis, Minnesota
| | - Loretta J Nastoupil
- Department of Lymphoma and Myeloma, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Olalekan O Oluwole
- Hematology Oncology and Stem Cell Transplant, Vanderbilt University Medical Center, Nashville, Tennessee
| | - Vivek G Patel
- Hematology Oncology and Stem Cell Transplant, Vanderbilt University Medical Center, Nashville, Tennessee
| | - Caspian Oliai
- Department of Medicine, UCLA David Geffen School of Medicine, Los Angeles, California
| | - Peter A Riedell
- The David and Etta Jonas Center for Cellular Therapy, University of Chicago, Chicago, Illinois
| | - Michael R Bishop
- The David and Etta Jonas Center for Cellular Therapy, University of Chicago, Chicago, Illinois
| | - Gunjan L Shah
- Department of Medicine, Adult Bone Marrow Transplant Service, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Miguel-Angel Perales
- Department of Medicine, Adult Bone Marrow Transplant Service, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Levanto Schachter
- Adult Blood and Marrow Stem Cell Transplant Program, Knight Cancer Institute, Oregon Health and Science University, Portland, Oregon
| | - Richard T Maziarz
- Adult Blood and Marrow Stem Cell Transplant Program, Knight Cancer Institute, Oregon Health and Science University, Portland, Oregon
| | | |
Collapse
|
219
|
Hao Y, Hsu WC, Parzynski CS, Degtyarev E, Hampson LV, Masood A, Wu WH. Effectiveness of tisagenlecleucel versus real-world standard of care in relapsed/refractory follicular lymphoma. J Comp Eff Res 2023; 12:e220173. [PMID: 37345672 PMCID: PMC10508300 DOI: 10.57264/cer-2022-0173] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2022] [Accepted: 04/06/2023] [Indexed: 06/23/2023] Open
Abstract
Aim: To contextualize the effectiveness of tisagenlecleucel versus real-world standard of care (SoC) in relapsed/refractory follicular lymphoma. Materials & methods: A retrospective indirect matched comparison study using data from the phase II ELARA trial and the US Flatiron Health Research Database. Results: Complete response rate was 69.1 versus 17.7% and the overall response rate was 85.6 versus 58.1% in tisagenlecleucel versus SoC, post weighting by odds. For overall survival, an estimated reduction in the risk of death was observed in favor of tisagenlecleucel over SoC. The hazard ratio for progression-free survival was 0.45 (95% CI: 0.26, 0.88), and for time-to-next treatment was 0.34 (95% CI: 0.15, 0.78) with tisagenlecleucel versus SoC. Conclusion: A consistent trend toward improved efficacy end points was observed in favor of tisagenlecleucel versus SoC.
Collapse
Affiliation(s)
- Yanni Hao
- Novartis Pharmaceuticals Corporation, East Hanover, NJ 07936, USA
- Current affiliation: Gilead Sciences, Foster City, CA 94404, USA
| | | | | | | | | | - Aisha Masood
- Novartis Pharmaceuticals Corporation, East Hanover, NJ 07936, USA
| | | |
Collapse
|
220
|
Saha A, Jhaveri K, Sarfraz H, Chavez JC. Tisagenlecleucel: CAR-T cell therapy for adult patients with relapsed or refractory follicular lymphoma. Expert Opin Biol Ther 2023; 23:869-876. [PMID: 37599463 DOI: 10.1080/14712598.2023.2248878] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2023] [Accepted: 08/14/2023] [Indexed: 08/22/2023]
Abstract
INTRODUCTION Tisagenlecleucel (tisa-cel) is an anti CD19 CAR-T therapy that has demonstrated clinical activity in R/R large B-cell lymphoma and R/R B-cell acute lymphoblastic leukemia. It showed particularly high efficacy in R/R follicular lymphoma (FL) with a manageable toxicity profile. The pivotal ELARA study in R/R FL confirmed these findings and led to the FDA approval of tisa-cel in R/R FL after two lines of systemic therapies. AREAS COVERED We start with an introduction of FL and the current treatment landscape with emphasis on the R/R setting. We review the role of CAR-T in R/R FL with focus on currently available products. We describe the ELARA study at a high level to give a perspective of the patient population that was treated. Finally, we discuss aspects related to product selection and whether bispecific antibodies will challenge the role of CAR-T in FL given their similar efficacy. EXPERT OPINION Tisa-cel is a highly effective therapy for heavily pretreated R/R FL with a toxicity profile that is low grade and manageable. Durable remissions (including high-risk patients) are seen in the pivotal ELARA study. Clinicians should consider early referral of R/R FL patients for assessment and discussion.
Collapse
Affiliation(s)
- Aditi Saha
- Department of Medicine/Hematology Oncology, University of South Florida, Tampa, FLUSA
| | - Khushali Jhaveri
- Department of Medicine/Hematology Oncology, University of South Florida, Tampa, FLUSA
| | - Humaira Sarfraz
- Department of Medicine/Hematology Oncology, University of South Florida, Tampa, FLUSA
| | - Julio C Chavez
- Department of Malignant Hematology, Moffitt Cancer Center, Tampa, FL USA
| |
Collapse
|
221
|
Watanabe T. Recent advances in treatment of nodal and gastrointestinal follicular lymphoma. World J Gastroenterol 2023; 29:3574-3594. [PMID: 37398889 PMCID: PMC10311612 DOI: 10.3748/wjg.v29.i23.3574] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/13/2023] [Revised: 05/14/2023] [Accepted: 05/22/2023] [Indexed: 06/16/2023] Open
Abstract
Follicular lymphoma (FL) is the most common low-grade lymphoma, and although nodal FL is highly responsive to treatment, the majority of patients relapse repeatedly, and the disease has been incurable with a poor prognosis. However, primary FL of the gastrointestinal tract has been increasingly detected in Japan, especially due to recent advances in small bowel endoscopy and increased opportunities for endoscopic examinations and endoscopic diagnosis. However, many cases are detected at an early stage, and the prognosis is good in many cases. In contrast, in Europe and the United States, gastrointestinal FL has long been considered to be present in 12%-24% of Stage-IV patients, and the number of advanced gastrointestinal cases is expected to increase. This editorial provides an overview of the recent therapeutic advances in nodal FL, including antibody-targeted therapy, bispecific antibody therapy, epigenetic modulation, and chimeric antigen receptor T-cell therapy, and reviews the latest therapeutic manuscripts published in the past year. Based on an understanding of the therapeutic advances in nodal FL, we also discuss future possibilities for gastroenterologists to treat gastrointestinal FL, especially in advanced cases.
Collapse
Affiliation(s)
- Takuya Watanabe
- Department of Internal Medicine and Gastroenterology, Watanabe Internal Medicine Aoyama Clinic, Niigata-city 9502002, Japan
| |
Collapse
|
222
|
Potnis KC, Huntington SF. Balancing considerations and qualifying conclusions for cost-effectiveness of therapies for relapsed/refractory follicular lymphoma. Blood Adv 2023; 7:2573-2574. [PMID: 36745098 PMCID: PMC10242627 DOI: 10.1182/bloodadvances.2023009864] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2023] [Accepted: 02/04/2023] [Indexed: 02/07/2023] Open
Affiliation(s)
| | - Scott F. Huntington
- Department of Internal Medicine, Yale School of Medicine, New Haven, CT
- Cancer Outcomes, Public Policy, and Effectiveness Research (COPPER) Center, Yale School of Medicine, New Haven, CT
| |
Collapse
|
223
|
Jiménez-Ubieto A, Martín-Muñoz A, Poza M, Dorado S, García-Ortiz A, Revilla E, Sarandeses P, Ruiz-Heredia Y, Baumann T, Rodríguez A, Calbacho M, Sánchez PM, Pina JMS, García-Sancho AM, Figaredo G, Gil-Alós D, Rufián L, Rodríguez M, Carneros L, Martínez-Laperche C, Bastos-Oreiro M, Wang C, Cedena MT, Rapado I, de Toledo P, Gallardo M, Valeri A, Ayala R, Martínez-López J, Barrio S. Personalized monitoring of circulating tumor DNA with a specific signature of trackable mutations after chimeric antigen receptor T-cell therapy in follicular lymphoma patients. Front Immunol 2023; 14:1188818. [PMID: 37342332 PMCID: PMC10277746 DOI: 10.3389/fimmu.2023.1188818] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2023] [Accepted: 04/26/2023] [Indexed: 06/22/2023] Open
Abstract
Background CART therapy has produced a paradigm shift in the treatment of relapsing FL patients. Strategies to optimize disease surveillance after these therapies are increasingly necessary. This study explores the potential value of ctDNA monitoring with an innovative signature of personalized trackable mutations. Method Eleven FL patients treated with anti-CD19 CAR T-cell therapy were included. One did not respond and was excluded. Genomic profiling was performed before starting lymphodepleting chemotherapy to identify somatic mutations suitable for LiqBio-MRD monitoring. The dynamics of the baseline mutations (4.5 per patient) were further analyzed on 59 cfDNA follow-up samples. PET/CT examinations were performed on days +90, +180, +365, and every six months until disease progression or death. Results After a median follow-up of 36 months, all patients achieved a CR as the best response. Two patients progressed. The most frequently mutated genes were CREBBP, KMT2D and EP300. Simultaneous analysis of ctDNA and PET/CT was available for 18 time-points. When PET/CT was positive, two out of four ctDNA samples were LiqBio-MRD negative. These two negative samples corresponded to women with a unique mesenteric mass in two evaluations and never relapsed. Meanwhile, 14 PET/CT negative images were mutation-free based on our LiqBio-MRD analysis (100%). None of the patients had a negative LiqBio-MRD test by day +7. Interestingly, all durably responding patients had undetectable ctDNA at or around three months after infusion. Two patients presented discordant results by PET/CT and ctDNA levels. No progression was confirmed in these cases. All the progressing patients were LiqBio-MRD positive before progression. Conclusion This is a proof-of-principle for using ctDNA to monitor response to CAR T-cell therapy in FL. Our results confirm that a non-invasive liquid biopsy MRD analysis may correlate with response and could be used to monitor response. Harmonized definitions of ctDNA molecular response and pinpointing the optimal timing for assessing ctDNA responses are necessary for this setting. If using ctDNA analysis, we suggest restricting follow-up PET/CT in CR patients to a clinical suspicion of relapse, to avoid false-positive results.
Collapse
Affiliation(s)
- Ana Jiménez-Ubieto
- Department of Hematology, Hospital Universitario 12 de Octubre, Instituto de Investigación Sanitaria Hospital 12 de Octubre (imas12), CNIO, CIBERONC, Madrid, Spain
| | - Alejandro Martín-Muñoz
- Department of Hematology, Hospital Universitario 12 de Octubre, Instituto de Investigación Sanitaria Hospital 12 de Octubre (imas12), CNIO, CIBERONC, Madrid, Spain
- Altum sequencing Co., Madrid, Spain
| | - María Poza
- Department of Hematology, Hospital Universitario 12 de Octubre, Instituto de Investigación Sanitaria Hospital 12 de Octubre (imas12), CNIO, CIBERONC, Madrid, Spain
| | - Sara Dorado
- Altum sequencing Co., Madrid, Spain
- Computational Science Department, Carlos III University, Madrid, Spain
| | - Almudena García-Ortiz
- Department of Hematology, Hospital Universitario 12 de Octubre, Instituto de Investigación Sanitaria Hospital 12 de Octubre (imas12), CNIO, CIBERONC, Madrid, Spain
| | - Enrique Revilla
- Departamento de Anatomía Patológica, Hospital Universitario 12 de Octubre, Madrid, Spain
| | - Pilar Sarandeses
- Departamento de Medicina Nuclear, Hospital Universitario 12 de Octubre, Madrid, Spain
| | - Yanira Ruiz-Heredia
- Department of Hematology, Hospital Universitario 12 de Octubre, Instituto de Investigación Sanitaria Hospital 12 de Octubre (imas12), CNIO, CIBERONC, Madrid, Spain
- Altum sequencing Co., Madrid, Spain
| | - Tycho Baumann
- Department of Hematology, Hospital Universitario 12 de Octubre, Instituto de Investigación Sanitaria Hospital 12 de Octubre (imas12), CNIO, CIBERONC, Madrid, Spain
| | - Antonia Rodríguez
- Department of Hematology, Hospital Universitario 12 de Octubre, Instituto de Investigación Sanitaria Hospital 12 de Octubre (imas12), CNIO, CIBERONC, Madrid, Spain
| | - María Calbacho
- Department of Hematology, Hospital Universitario 12 de Octubre, Instituto de Investigación Sanitaria Hospital 12 de Octubre (imas12), CNIO, CIBERONC, Madrid, Spain
| | - Pilar Martínez Sánchez
- Department of Hematology, Hospital Universitario 12 de Octubre, Instituto de Investigación Sanitaria Hospital 12 de Octubre (imas12), CNIO, CIBERONC, Madrid, Spain
| | - José María Sánchez Pina
- Department of Hematology, Hospital Universitario 12 de Octubre, Instituto de Investigación Sanitaria Hospital 12 de Octubre (imas12), CNIO, CIBERONC, Madrid, Spain
| | | | - Gloria Figaredo
- Department of Hematology, Hospital Universitario 12 de Octubre, Instituto de Investigación Sanitaria Hospital 12 de Octubre (imas12), CNIO, CIBERONC, Madrid, Spain
| | - Daniel Gil-Alós
- Department of Hematology, Hospital Universitario 12 de Octubre, Instituto de Investigación Sanitaria Hospital 12 de Octubre (imas12), CNIO, CIBERONC, Madrid, Spain
| | - Laura Rufián
- Department of Hematology, Hospital Universitario 12 de Octubre, Instituto de Investigación Sanitaria Hospital 12 de Octubre (imas12), CNIO, CIBERONC, Madrid, Spain
- Altum sequencing Co., Madrid, Spain
| | - Margarita Rodríguez
- Department of Hematology, Hospital Universitario 12 de Octubre, Instituto de Investigación Sanitaria Hospital 12 de Octubre (imas12), CNIO, CIBERONC, Madrid, Spain
- Altum sequencing Co., Madrid, Spain
| | - Laura Carneros
- Department of Hematology, Hospital Universitario 12 de Octubre, Instituto de Investigación Sanitaria Hospital 12 de Octubre (imas12), CNIO, CIBERONC, Madrid, Spain
| | | | | | - Chongwu Wang
- Hosea Precision Medical Technology Co., Ltd., Weihai, Shangdong, China
| | - María-Teresa Cedena
- Department of Hematology, Hospital Universitario 12 de Octubre, Instituto de Investigación Sanitaria Hospital 12 de Octubre (imas12), CNIO, CIBERONC, Madrid, Spain
| | - Inmaculada Rapado
- Department of Hematology, Hospital Universitario 12 de Octubre, Instituto de Investigación Sanitaria Hospital 12 de Octubre (imas12), CNIO, CIBERONC, Madrid, Spain
| | - Paula de Toledo
- Computational Science Department, Carlos III University, Madrid, Spain
| | - Miguel Gallardo
- Department of Hematology, Hospital Universitario 12 de Octubre, Instituto de Investigación Sanitaria Hospital 12 de Octubre (imas12), CNIO, CIBERONC, Madrid, Spain
| | - Antonio Valeri
- Department of Hematology, Hospital Universitario 12 de Octubre, Instituto de Investigación Sanitaria Hospital 12 de Octubre (imas12), CNIO, CIBERONC, Madrid, Spain
| | - Rosa Ayala
- Department of Hematology, Hospital Universitario 12 de Octubre, Instituto de Investigación Sanitaria Hospital 12 de Octubre (imas12), CNIO, CIBERONC, Madrid, Spain
| | - Joaquín Martínez-López
- Department of Hematology, Hospital Universitario 12 de Octubre, Instituto de Investigación Sanitaria Hospital 12 de Octubre (imas12), CNIO, CIBERONC, Madrid, Spain
| | - Santiago Barrio
- Department of Hematology, Hospital Universitario 12 de Octubre, Instituto de Investigación Sanitaria Hospital 12 de Octubre (imas12), CNIO, CIBERONC, Madrid, Spain
- Altum sequencing Co., Madrid, Spain
| |
Collapse
|
224
|
Friedberg JW. Update on follicular lymphoma. Hematol Oncol 2023; 41 Suppl 1:43-47. [PMID: 37294960 PMCID: PMC10264144 DOI: 10.1002/hon.3138] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2023] [Accepted: 03/28/2023] [Indexed: 06/11/2023]
Abstract
The past two decades have seen remarkable progress in both biological understanding and optimizing treatment of follicular lymphoma. Historically considered an incurable disease, long-term follow-up of several induction approaches demonstrates that up to 40% of patients enjoy remission durations of 10 or more years, and risk of dying of lymphoma continues to fall. This update will focus on progress in follicular lymphoma over the past 3 years, which has included refinements in staging and prognosis, novel immunotherapy treatment approaches for relapsed and refractory disease, and long-term follow-up of pivotal trials. Ongoing trials will define the optimal sequence for these novel treatments, including whether earlier incorporation of these approaches may result in definitive cure of this disease. Through ongoing and planned correlative studies, we are poised to ultimately achieve the goal of a precision management approach to follicular lymphoma.
Collapse
|
225
|
Chatwal MS, Chahoud J, Spiess PE. Revisiting mechanisms of resistance to immunotherapies in metastatic clear-cell renal-cell carcinoma. CANCER DRUG RESISTANCE (ALHAMBRA, CALIF.) 2023; 6:314-326. [PMID: 37457132 PMCID: PMC10344725 DOI: 10.20517/cdr.2023.09] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 02/11/2023] [Revised: 05/02/2023] [Accepted: 05/25/2023] [Indexed: 07/18/2023]
Abstract
Renal-cell carcinoma (RCC) remains a leading cause of cancer-related mortality worldwide. Though newer therapeutic combinations of immune checkpoint inhibitors and targeted therapies have greatly improved outcomes, resistance to these therapies is becoming a challenge for long-term control. Mechanisms of resistance have been explored in a variety of solid tumors, including RCC. Based upon our review of the current literature on the mechanisms of resistance to immunotherapies for the management of metastatic clear-cell renal cell carcinomas (mccRCC), the ensuing conclusions have been made: The management of mccRCC has progressed substantially with the advent of checkpoint inhibitors and targeted oral therapies, alone and/or in combination. Nevertheless, innate or developed resistance to these therapies remains an ongoing challenge, particularly to immune checkpoint inhibitors (ICIs). Several of the known mechanisms of resistance have been well defined, but recent progression in cellular therapies helps to expand the armamentarium of potential combination options that may overcome these modes of resistance and improve long-term disease control and survival for an otherwise dismal disease. In the ensuing review and update of the literature on the mechanisms of resistance to immunotherapies in mccRCC, we have revisited the known resistance mechanisms of immunotherapies in metastatic clear-cell RCC and explored ongoing and future strategies to overcome them.
Collapse
Affiliation(s)
- Monica Sheila Chatwal
- Correspondence to: Dr. Monica Sheila Chatwal, Department of Genitourinary Oncology, H. Lee Moffitt Cancer Center and Research Institute, 12902 USF Magnolia Drive, Tampa, FL33612, USA. E-mail:
| | | | | |
Collapse
|
226
|
Huang S, Wang X, Wang Y, Wang Y, Fang C, Wang Y, Chen S, Chen R, Lei T, Zhang Y, Xu X, Li Y. Deciphering and advancing CAR T-cell therapy with single-cell sequencing technologies. Mol Cancer 2023; 22:80. [PMID: 37149643 PMCID: PMC10163813 DOI: 10.1186/s12943-023-01783-1] [Citation(s) in RCA: 9] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2023] [Accepted: 04/26/2023] [Indexed: 05/08/2023] Open
Abstract
Chimeric antigen receptor (CAR) T-cell therapy has made remarkable progress in cancer immunotherapy, but several challenges with unclear mechanisms hinder its wide clinical application. Single-cell sequencing technologies, with the powerful unbiased analysis of cellular heterogeneity and molecular patterns at unprecedented resolution, have greatly advanced our understanding of immunology and oncology. In this review, we summarize the recent applications of single-cell sequencing technologies in CAR T-cell therapy, including the biological characteristics, the latest mechanisms of clinical response and adverse events, promising strategies that contribute to the development of CAR T-cell therapy and CAR target selection. Generally, we propose a multi-omics research mode to guide potential future research on CAR T-cell therapy.
Collapse
Affiliation(s)
- Shengkang Huang
- The Second School of Clinical Medicine, Zhujiang Hospital, Southern Medical University, Guangzhou, China
- State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Xinyu Wang
- The Second School of Clinical Medicine, Zhujiang Hospital, Southern Medical University, Guangzhou, China
| | - Yu Wang
- The First School of Clinical Medicine, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Yajing Wang
- The Second School of Clinical Medicine, Zhujiang Hospital, Southern Medical University, Guangzhou, China
| | - Chenglong Fang
- The Second School of Clinical Medicine, Zhujiang Hospital, Southern Medical University, Guangzhou, China
| | - Yazhuo Wang
- The Second School of Clinical Medicine, Zhujiang Hospital, Southern Medical University, Guangzhou, China
- School of Rehabilitation Sciences, Southern Medical University, Guangzhou, China
| | - Sifei Chen
- The Second School of Clinical Medicine, Zhujiang Hospital, Southern Medical University, Guangzhou, China
- State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Runkai Chen
- The Second School of Clinical Medicine, Zhujiang Hospital, Southern Medical University, Guangzhou, China
| | - Tao Lei
- The Second School of Clinical Medicine, Zhujiang Hospital, Southern Medical University, Guangzhou, China
| | - Yuchen Zhang
- The Second School of Clinical Medicine, Zhujiang Hospital, Southern Medical University, Guangzhou, China
| | - Xinjie Xu
- State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China.
| | - Yuhua Li
- Department of Hematology, Zhujiang Hospital, Southern Medical University, Guangzhou, China.
- Bioland Laboratory (Guangzhou Regenerative Medicine and Health Guangdong Laboratory), Guangzhou, China.
| |
Collapse
|
227
|
Rivero A, Mozas P, Magnano L, López-Guillermo A. Novel targeted drugs for follicular and marginal zone lymphoma: a comprehensive review. Front Oncol 2023; 13:1170394. [PMID: 37207160 PMCID: PMC10189145 DOI: 10.3389/fonc.2023.1170394] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2023] [Accepted: 04/03/2023] [Indexed: 05/21/2023] Open
Abstract
Although mostly incurable, indolent non-Hodgkin lymphomas (iNHL) are chronic diseases with a median overall survival approaching 20 years. In recent years, important advances in the knowledge of the biology of these lymphomas have led to the development of new drugs, mostly chemotherapy-free, with promising outcomes. With a median age of around 70 years at diagnosis, many patients with iNHL suffer from comorbid conditions that may limit treatment options. Therefore, nowadays, in the transition towards personalized medicine, several challenges lie ahead, such as identifying predictive markers for the selection of treatment, the adequate sequencing of available therapies, and the management of new and accumulated toxicities. In this review, we include a perspective on recent therapeutic advances in follicular and marginal zone lymphoma. We describe emerging data on approved and emerging novel therapies, such as targeted therapies (PI3K inhibitors, BTK inhibitors, EZH2 inhibitors), monoclonal antibodies and antibody-drug conjugates. Finally, we describe immune-directed approaches such as combinations with lenalidomide or the even more innovative bispecific T-cell engagers and chimeric antigen receptor T-cell therapy, which can achieve a high rate of durable responses with manageable toxicities, further obviating the need for chemotherapy.
Collapse
Affiliation(s)
- Andrea Rivero
- Department of Hematology, Hospital Clínic de Barcelona, Barcelona, Spain
- Institut d’Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, Spain
| | - Pablo Mozas
- Department of Hematology, Hospital Clínic de Barcelona, Barcelona, Spain
- Institut d’Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, Spain
| | - Laura Magnano
- Department of Hematology, Hospital Clínic de Barcelona, Barcelona, Spain
- Institut d’Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, Spain
| | - Armando López-Guillermo
- Department of Hematology, Hospital Clínic de Barcelona, Barcelona, Spain
- Institut d’Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, Spain
- Universitat de Barcelona, Barcelona, Spain
| |
Collapse
|
228
|
Alderuccio JP, Lossos IS. Enhancing prognostication and personalizing treatment of extranodal marginal zone lymphoma. Expert Rev Hematol 2023; 16:333-348. [PMID: 37086394 PMCID: PMC10183153 DOI: 10.1080/17474086.2023.2206557] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2023] [Accepted: 04/20/2023] [Indexed: 04/23/2023]
Abstract
INTRODUCTION Extranodal marginal zone lymphoma (EMZL) of mucosa-associated lymphoid tissue is an indolent lymphoma originating from marginal zone B-cells and associated with chronic inflammation. EMZL demonstrates distinct genomic alterations according to the primary extranodal site of disease but commonly affects signaling pathways including NF-ĸB, B-cell receptor, and NOTCH. Treatment with radiation therapy is commonly implemented in localized diseases, and multiple agents are available for patients with advanced-stage diseases in need of therapy. Bendamustine with rituximab is a frontline platform associated with high efficacy. AREAS COVERED Clinical features, diagnosis, genomics, models enabling risk stratification, treatment options, and future directions. EXPERT OPINION The lack of consistent genotyping profile in EMZL precludes the development of tissue and circulatory biomarkers for the diagnosis, risk stratification, and monitoring of minimal residual disease. Furthermore, the biological heterogeneity observed in extranodal sites associated with overall limited genomic data prevents the testing of druggable pathways aiming for a personalized treatment approach. Future clinical trials should focus on EMZL considering the unique clinical characteristics in the eligibility criteria and response assessment to better inform efficacy of novel agents and delineate sequences of therapies.
Collapse
Affiliation(s)
| | - Izidore S. Lossos
- Department of Medicine, Division of Hematology
- Department of Molecular and Cellular Pharmacology, Sylvester Comprehensive Cancer Center, University of Miami Miller School of Medicine, Miami, FL, USA
| |
Collapse
|
229
|
Zanoni L, Bezzi D, Nanni C, Paccagnella A, Farina A, Broccoli A, Casadei B, Zinzani PL, Fanti S. PET/CT in Non-Hodgkin Lymphoma: An Update. Semin Nucl Med 2023; 53:320-351. [PMID: 36522191 DOI: 10.1053/j.semnuclmed.2022.11.001] [Citation(s) in RCA: 8] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2022] [Revised: 11/09/2022] [Accepted: 11/10/2022] [Indexed: 12/15/2022]
Abstract
Non-Hodgkin lymphomas represents a heterogeneous group of lymphoproliferative disorders characterized by different clinical courses, varying from indolent to highly aggressive. 18F-FDG-PET/CT is the current state-of-the-art diagnostic imaging, for the staging, restaging and evaluation of response to treatment in lymphomas with avidity for 18F-FDG, despite it is not routinely recommended for surveillance. PET-based response criteria (using five-point Deauville Score) are nowadays uniformly applied in FDG-avid lymphomas. In this review, a comprehensive overview of the role of 18F-FDG-PET in Non-Hodgkin lymphomas is provided, at each relevant point of patient management, particularly focusing on recent advances on diffuse large B-cell lymphoma and follicular lymphoma, with brief updates also on other histotypes (such as marginal zone, mantle cell, primary mediastinal- B cell lymphoma and T cell lymphoma). PET-derived semiquantitative factors useful for patient stratification and prognostication and emerging radiomics research are also presented.
Collapse
Affiliation(s)
- Lucia Zanoni
- Nuclear Medicine, IRCCS Azienda Ospedaliero-Universitaria di Bologna, Bologna, Italy.
| | - Davide Bezzi
- Nuclear Medicine, Alma Mater Studiorum University of Bologna, Bologna, Italy
| | - Cristina Nanni
- Nuclear Medicine, IRCCS Azienda Ospedaliero-Universitaria di Bologna, Bologna, Italy
| | - Andrea Paccagnella
- Nuclear Medicine, Alma Mater Studiorum University of Bologna, Bologna, Italy; Nuclear Medicine Unit, AUSL Romagna, Cesena, Italy
| | - Arianna Farina
- Nuclear Medicine, Alma Mater Studiorum University of Bologna, Bologna, Italy
| | - Alessandro Broccoli
- IRCCS Azienda Ospedaliero-Universitaria di Bologna, Istituto di Ematologia "Seràgnoli," Bologna, Italy; Dipartimento di Medicina Specialistica, Diagnostica e Sperimentale, Università di Bologna, Bologna, Italy
| | - Beatrice Casadei
- IRCCS Azienda Ospedaliero-Universitaria di Bologna, Istituto di Ematologia "Seràgnoli," Bologna, Italy; Dipartimento di Medicina Specialistica, Diagnostica e Sperimentale, Università di Bologna, Bologna, Italy
| | - Pier Luigi Zinzani
- IRCCS Azienda Ospedaliero-Universitaria di Bologna, Istituto di Ematologia "Seràgnoli," Bologna, Italy; Dipartimento di Medicina Specialistica, Diagnostica e Sperimentale, Università di Bologna, Bologna, Italy
| | - Stefano Fanti
- Nuclear Medicine, IRCCS Azienda Ospedaliero-Universitaria di Bologna, Bologna, Italy; Nuclear Medicine, Alma Mater Studiorum University of Bologna, Bologna, Italy
| |
Collapse
|
230
|
Bogatu A, Wysocka M, Wysocki O, Butterworth H, Pillai M, Allison J, Landers D, Kilgour E, Thistlethwaite F, Freitas A. Meta-analysis informed machine learning: Supporting cytokine storm detection during CAR-T cell Therapy. J Biomed Inform 2023; 142:104367. [PMID: 37105509 DOI: 10.1016/j.jbi.2023.104367] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2022] [Revised: 04/11/2023] [Accepted: 04/17/2023] [Indexed: 04/29/2023]
Abstract
Cytokine release syndrome (CRS), also known as cytokine storm, is one of the most consequential adverse effects of chimeric antigen receptor therapies that have shown otherwise promising results in cancer treatment. When emerging, CRS could be identified by the analysis of specific cytokine and chemokine profiles that tend to exhibit similarities across patients. In this paper, we exploit these similarities using machine learning algorithms and set out to pioneer a meta-review informed method for the identification of CRS based on specific cytokine peak concentrations and evidence from previous clinical studies. To this end we also address a widespread challenge of the applicability of machine learning in general: reduced training data availability. We do so by augmenting available (but often insufficient) patient cytokine concentrations with statistical knowledge extracted from domain literature. We argue that such methods could support clinicians in analyzing suspect cytokine profiles by matching them against the said CRS knowledge from past clinical studies, with the ultimate aim of swift CRS diagnosis. We evaluate our proposed methods under several design choices, achieving performance of more than 90% in terms of CRS identification accuracy, and showing that many of our choices outperform a purely data-driven alternative. During evaluation with real-world CRS clinical data, we emphasize the potential of our proposed method of producing interpretable results, in addition to being effective in identifying the onset of cytokine storm.
Collapse
Affiliation(s)
- Alex Bogatu
- Department of Computer Science, The University of Manchester, United Kingdom; Digital Experimental Cancer Medicine Team, Cancer Biomarker Centre, CRUK Manchester Institute, United Kingdom.
| | - Magdalena Wysocka
- Digital Experimental Cancer Medicine Team, Cancer Biomarker Centre, CRUK Manchester Institute, United Kingdom
| | - Oskar Wysocki
- Department of Computer Science, The University of Manchester, United Kingdom; Digital Experimental Cancer Medicine Team, Cancer Biomarker Centre, CRUK Manchester Institute, United Kingdom
| | | | - Manon Pillai
- Division of Cancer Sciences, Faculty of Biology, Medicine and Health, The University of Manchester, UK
| | - Jennifer Allison
- Division of Cancer Sciences, Faculty of Biology, Medicine and Health, The University of Manchester, UK
| | - Dónal Landers
- Digital Experimental Cancer Medicine Team, Cancer Biomarker Centre, CRUK Manchester Institute, United Kingdom
| | - Elaine Kilgour
- Cancer Biomarker Centre, CRUK Manchester Institute, United Kingdom
| | - Fiona Thistlethwaite
- Division of Cancer Sciences, Faculty of Biology, Medicine and Health, The University of Manchester, UK; The Christie NHS Foundation Trust, Manchester, UK
| | - André Freitas
- Department of Computer Science, The University of Manchester, United Kingdom; Digital Experimental Cancer Medicine Team, Cancer Biomarker Centre, CRUK Manchester Institute, United Kingdom; Idiap Research Institute, Switzerland
| |
Collapse
|
231
|
Cappell KM, Kochenderfer JN. Long-term outcomes following CAR T cell therapy: what we know so far. Nat Rev Clin Oncol 2023; 20:359-371. [PMID: 37055515 PMCID: PMC10100620 DOI: 10.1038/s41571-023-00754-1] [Citation(s) in RCA: 314] [Impact Index Per Article: 314.0] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/14/2023] [Indexed: 04/15/2023]
Abstract
Chimeric antigen receptors (CAR) are engineered fusion proteins designed to target T cells to antigens expressed on cancer cells. CAR T cells are now an established treatment for patients with relapsed and/or refractory B cell lymphomas, B cell acute lymphoblastic leukaemia and multiple myeloma. At the time of this writing, over a decade of follow-up data are available from the initial patients who received CD19-targeted CAR T cells for B cell malignancies. Data on the outcomes of patients who received B cell maturation antigen (BCMA)-targeted CAR T cells for multiple myeloma are more limited owing to the more recent development of these constructs. In this Review, we summarize long-term follow-up data on efficacy and toxicities from patients treated with CAR T cells targeting CD19 or BCMA. Overall, the data demonstrate that CD19-targeted CAR T cells can induce prolonged remissions in patients with B cell malignancies, often with minimal long-term toxicities, and are probably curative for a subset of patients. By contrast, remissions induced by BCMA-targeted CAR T cells are typically more short-lived but also generally have only limited long-term toxicities. We discuss factors associated with long-term remissions, including the depth of initial response, malignancy characteristics predictive of response, peak circulating CAR levels and the role of lymphodepleting chemotherapy. We also discuss ongoing investigational strategies designed to improve the length of remission following CAR T cell therapy.
Collapse
Affiliation(s)
- Kathryn M Cappell
- Surgery Branch, Center for Cancer Research, National Cancer Institute (NCI), NIH, Bethesda, MD, USA
| | - James N Kochenderfer
- Surgery Branch, Center for Cancer Research, National Cancer Institute (NCI), NIH, Bethesda, MD, USA.
| |
Collapse
|
232
|
Velasco R, Mussetti A, Villagrán-García M, Sureda A. CAR T-cell-associated neurotoxicity in central nervous system hematologic disease: Is it still a concern? Front Neurol 2023; 14:1144414. [PMID: 37090983 PMCID: PMC10117964 DOI: 10.3389/fneur.2023.1144414] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2023] [Accepted: 03/06/2023] [Indexed: 04/25/2023] Open
Abstract
Chimeric antigen receptor (CAR) T-cell systemic immunotherapy has revolutionized how clinicians treat several refractory and relapsed hematologic malignancies. Due to its peculiar mechanism of action, CAR T-cell-based therapy has enlarged the spectrum of neurological toxicities. CAR T-cell-associated neurotoxicity-initially defined as CAR T-cell-related encephalopathy syndrome (CRES) and currently coined within the acronym ICANS (immune effector cell-associated neurotoxicity syndrome)-is perhaps the most concerning toxicity of CAR T-cell therapy. Importantly, hematologic malignancies (especially lymphoid malignancies) may originate in or spread to the central nervous system (CNS) in the form of parenchymal and/or meningeal disease. Due to the emergence of deadly and neurological adverse events, such as fatal brain edema in some patients included in early CAR T-cell trials, safety concerns for those with CNS primary or secondary infiltration arose and contributed to the routine exclusion of individuals with pre-existing or active CNS involvement from pivotal trials. However, based primarily on the lack of evidence, it remains unknown whether CNS involvement increases the risk and/or severity of CAR T-cell-related neurotoxicity. Given the limited treatment options available for patients once they relapse with CNS involvement, it is of high interest to explore the role of novel clinical strategies including CAR T cells to treat leukemias/lymphomas and myeloma with CNS involvement. The purpose of this review was to summarize currently available neurological safety data of CAR T-cell-based immunotherapy from the clinical trials and real-world experiences in adult patients with CNS disease due to lymphoma, leukemia, or myeloma. Increasing evidence supports that CNS involvement in hematologic disease should no longer be considered per se as an absolute contraindication to CAR T-cell-based therapy. While the incidence may be high, severity does not appear to be impacted significantly by pre-existing CNS status. Close monitoring by trained neurologists is recommended.
Collapse
Affiliation(s)
- Roser Velasco
- Neuro-Oncology Unit, Department of Neurology, Hospital Universitari de Bellvitge-Institut Català d'Oncologia, Barcelona, Spain
- Department of Cell Biology, Physiology and Immunology, Institute of Neurosciences, Cerdanyola del Vallés, Spain
| | - Alberto Mussetti
- Department of Hematology, Catalan Institute of Oncology, Hospital Duran i Reynals, Institut d'Investigació Biomèdica de Bellvitge (IDIBELL), L'Hospitalet de Llobregat, Barcelona, Spain
| | - Macarena Villagrán-García
- French Reference Center on Paraneoplastic Neurological Syndromes and Autoimmune Encephalitis, Hospices Civils de Lyon, Hôpital Neurologique, Bron. UMR MeLiS team SynatAc, INSERM1314/CNRS5284, Lyon, France
| | - Anna Sureda
- Department of Hematology, Catalan Institute of Oncology, Hospital Duran i Reynals, Institut d'Investigació Biomèdica de Bellvitge (IDIBELL), L'Hospitalet de Llobregat, Barcelona, Spain
- Medicine Department, Universitat de Barcelona, Barcelona, Spain
| |
Collapse
|
233
|
Chohan KL, Siegler EL, Kenderian SS. CAR-T Cell Therapy: the Efficacy and Toxicity Balance. Curr Hematol Malig Rep 2023; 18:9-18. [PMID: 36763238 PMCID: PMC10505056 DOI: 10.1007/s11899-023-00687-7] [Citation(s) in RCA: 23] [Impact Index Per Article: 23.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 12/20/2022] [Indexed: 02/11/2023]
Abstract
PURPOSE OF REVIEW Chimeric antigen receptor (CAR) T cell therapy is an immunotherapy that has resulted in tremendous progress in the treatment of patients with B cell malignancies. However, the remarkable efficacy of therapy is not without significant safety concerns. Herein, we will review the unique and potentially life-threatening toxicities associated with CAR-T cell therapy and their association with treatment efficacy. RECENT FINDINGS Currently, CAR-T cell therapy is approved for the treatment of B cell relapsed or refractory leukemia and lymphoma, and most recently, multiple myeloma (MM). In these different diseases, it has led to excellent complete and overall response rates depending on the patient population and therapy. Despite promising efficacy, CAR-T cell therapy is associated with significant side effects; the two most notable toxicities are cytokine release syndrome (CRS) and immune effector cell-associated neurotoxicity syndrome (ICANS). The treatment of CAR-T-induced toxicity is supportive; however, as higher-grade adverse events occur, toxicity-directed therapy with tocilizumab, an IL-6 receptor antibody, and steroids is standard practice. Overall, a careful risk-benefit balance exists between the efficacy and toxicities of therapies. The challenge lies in the underlying pathophysiology of CAR-T-related toxicity which relies upon the activation of CAR-T cells. Some degree of toxicity is expected to achieve an effective response to therapy, and certain aspects of treatment are also associated with toxicity. As progress is made in the investigation and approval of new CARs, novel toxicity-directed therapies and toxicity-limited constructs will be the focus of attention.
Collapse
Affiliation(s)
| | - Elizabeth L Siegler
- T Cell Engineering, Mayo Clinic, Rochester, MN, USA
- Division of Hematology, Mayo Clinic, Rochester, MN, USA
| | - Saad S Kenderian
- T Cell Engineering, Mayo Clinic, Rochester, MN, USA.
- Division of Hematology, Mayo Clinic, Rochester, MN, USA.
- Department of Molecular Medicine, Mayo Clinic, Rochester, MN, USA.
- Department of Immunology, Mayo Clinic, Rochester, MN, USA.
- Department of Molecular Pharmacology & Experimental Therapeutics, Mayo Clinic, 200 1st ST SW, Rochester, MN, 55902, USA.
| |
Collapse
|
234
|
Cellular Therapies in Chronic Lymphocytic Leukemia and Richter’s Transformation: Recent Developments in Chimeric Antigen Receptor T-Cells, Natural Killer Cells, and Allogeneic Stem Cell Transplant. Cancers (Basel) 2023; 15:cancers15061838. [PMID: 36980726 PMCID: PMC10046903 DOI: 10.3390/cancers15061838] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2023] [Revised: 03/09/2023] [Accepted: 03/14/2023] [Indexed: 03/22/2023] Open
Abstract
Cellular therapies can be viewed as both the newest and oldest techniques for treating chronic lymphocytic leukemia (CLL) and Richter’s transformation (RT). On one hand, allogeneic hematopoietic stem cell transplantation (alloHSCT) has been available for decades, though its use is diminishing with the increasing availability of effective novel targeted agents, especially in CLL. Among newer techniques, chimeric antigen receptor T-cells (CAR-T) have demonstrated astounding efficacy in several hematologic malignancies, leading to FDA approval and use in clinical practice. However, though CLL is the earliest disease type for which CAR-T were studied, development has been slower and has yet to lead to regulatory approval. Owing partially to its rarity but also due to the aggressive behavior of RT, CAR-T in RT have only been minimally explored. Here, we will focus on the applications of cellular therapies in CLL and RT, specifically reviewing more recent data related to alloHSCT in the novel-agent era and CAR-T cell development in CLL/RT, focusing on safety and efficacy successes and limitations. We will review strategies to improve upon CAR-T efficacy and discuss ongoing trials utilizing CAR-T in CLL/RT, as well as emerging technologies, such as allogeneic CAR-T and natural killer CAR (CAR NK) cells.
Collapse
|
235
|
Eyre TA, Barrington SF, Okosun J, Abamba C, Pearce RM, Lee J, Carpenter B, Crawley CR, Bloor AJC, Gilleece M, Nicholson E, Shah N, Orchard K, Malladi R, Townsend WM. Impact of positron emission tomography - computed tomography status on progression-free survival for relapsed follicular lymphoma patients undergoing autologous stem cell transplantation. Haematologica 2023; 108:785-796. [PMID: 35586966 PMCID: PMC9973492 DOI: 10.3324/haematol.2021.280287] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2021] [Accepted: 04/28/2022] [Indexed: 11/09/2022] Open
Abstract
The optimum management approach for patients with relapsed or refractory follicular lymphoma remains uncertain. Autologous stem cell transplantation (autoSCT) is considered a standard option in suitable, younger patients with relapsed follicular lymphoma. AutoSCT is associated with very durable remissions in a minority of subjects, but also with significant, well-established toxicities. Although positron emission tomography (PET) status prior to autoSCT is an established prognostic factor in diffuse large B-cell lymphoma and Hodgkin lymphoma, no data exist in follicular lymphoma. We describe survival outcomes according to pre-transplant PET status, classified by the Lugano criteria into complete metabolic remission (CMR) versus non-CMR, in 172 patients with relapsed or refractory follicular lymphoma within a national, multicenter, retrospective British Society of Blood and Marrow Transplantation and Cellular Therapy registry study. The median number of lines of therapy prior to SCT was three (range, 1-6). The median follow-up after SCT was 27 months (range, 3-70). The median progression-free survival for all patients after autoSCT was 28 months (interquartile range, 23- 36). There was no interaction between age at transplantation, sex, number of months since last relapse, Karnofsky performance status or comorbidity index and achieving CMR prior to autoSCT. Superior progression-free survival was observed in 115 (67%) patients obtaining CMR versus 57 (33%) non-CMR patients (3-year progression-free survival 50% vs. 22%, P=0.011) and by pre-SCT Deauville score (continuous variable 1-5, hazard ratio [HR]=1.32, P=0.049). PET status was independently associated with progression-free status (non-CMR HR=2.02, P=0.003), overall survival (non-CMR HR=3.08, P=0.010) and risk of relapse (non-CMR HR=1.64, P=0.046) after autoSCT by multivariable analysis. Our data suggest that pre- SCT PET status is of clear prognostic value and may help to improve the selection of patients for autoSCT.
Collapse
Affiliation(s)
- Toby A Eyre
- Department of Haematology, Churchill Hospital, Oxford University Hospitals NHS Foundation Trust, Oxford.
| | - Sally F Barrington
- King's College London and Guy's and St Thomas' PET Centre, School of Biomedical Engineering and Imaging Sciences, King's College London, King's Health Partners, London
| | - Jessica Okosun
- Centre for Haemato-Oncology, Barts Cancer Institute, Queen Mary University of London, London
| | - Clementina Abamba
- BSBMTCT data registry, 5th Floor Tabard House, Talbot Yard, Guy's Hospital, Great Maze Pond, London
| | - Rachel M Pearce
- BSBMTCT data registry, 5th Floor Tabard House, Talbot Yard, Guy's Hospital, Great Maze Pond, London
| | - Julia Lee
- Centre for Haemato-Oncology, Barts Cancer Institute, Queen Mary University of London, London
| | - Ben Carpenter
- Department of Haematology, University College London Hospitals 235 Euston Road, London
| | - Charles R Crawley
- Department of Haematology and Bone Marrow Transplantation, Addenbrookes Hospital, Cambridge
| | - Adrian J C Bloor
- Department of Haematology, The Christie Hospital NHS Trust, Manchester
| | - Maria Gilleece
- Department of Haematology and Bone Marrow Transplantation, Leeds Teaching Hospitals NHS Trust, Leeds
| | - Emma Nicholson
- Department of Haematology and Bone Marrow Transplantation, Royal Marsden Hospital, London
| | - Nimish Shah
- Department of Haematology, Norfolk and Norwich University Hospitals, Norwich
| | - Kim Orchard
- Department of Haematology and Bone Marrow Transplantation, Southampton University Hospitals, Southampton
| | - Ram Malladi
- Department of Haematology and Bone Marrow Transplantation, Addenbrookes Hospital, Cambridge
| | - William M Townsend
- Department of Haematology, University College London Hospitals 235 Euston Road, London
| |
Collapse
|
236
|
Chu Y, Gardenswartz A, Diorio C, Marks LJ, Lowe E, Teachey DT, Cairo MS. Cellular and humoral immunotherapy in children, adolescents and young adults with non-Hodgkin lymphoma. Best Pract Res Clin Haematol 2023; 36:101442. [PMID: 36907635 DOI: 10.1016/j.beha.2023.101442] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2023] [Accepted: 01/31/2023] [Indexed: 02/05/2023]
Abstract
The prognosis is dismal (2-year overall survival less than 25%) for childhood, adolescent, and young adult (CAYA) with relapsed and/or refractory (R/R) non-Hodgkin lymphoma (NHL). Novel targeted therapies are desperately needed for this poor-risk population. CD19, CD20, CD22, CD79a, CD38, CD30, LMP1 and LMP2 are attractive targets for immunotherapy in CAYA patients with R/R NHL. Novel anti-CD20 monoclonal antibodies, anti-CD38 monoclonal antibody, antibody drug conjugates and T and natural killer (NK)-cell bispecific and trispecific engagers are being investigated in the R/R setting and are changing the landscape of NHL therapy. A variety of cellular immunotherapies such as viral activated cytotoxic T-lymphocyte, chimeric antigen receptor (CAR) T-cells, NK and CAR NK-cells have been investigated and provide alternative options for CAYA patients with R/R NHL. Here, we provide an update and clinical practice guidance of utilizing these cellular and humoral immunotherapies in CAYA patients with R/R NHL.
Collapse
Affiliation(s)
- Yaya Chu
- Department of Pediatrics, New York Medical College, Valhalla, NY, USA
| | | | - Caroline Diorio
- The Children's Hospital of Philadelphia, Philadelphia, PA, USA
| | - Lianna J Marks
- Division of Pediatric Hematology and Oncology, Stanford University School of Medicine, Palo Alto, CA, USA
| | - Eric Lowe
- Division of Pediatric Hematology-Oncology, Children's Hospital of the Kings Daughter, Norfolk, VA, USA
| | - David T Teachey
- The Children's Hospital of Philadelphia, Philadelphia, PA, USA; University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
| | - Mitchell S Cairo
- Department of Pediatrics, New York Medical College, Valhalla, NY, USA; Department of Epidemiology and Community Health, New York Medical College, Valhalla, NY, USA; Department of Medicine, New York Medical College, Valhalla, NY, USA; Department of Pathology, Microbiology and Immunology, New York Medical College, Valhalla, NY, USA; Department of Cell Biology, New York Medical College, Valhalla, NY, USA; Department of Anatomy, New York Medical College, Valhalla, NY, USA.
| |
Collapse
|
237
|
Sarkozy C, Salles G. Positron emission tomography-computed tomography before autologous stem cell transplant in follicular lymphoma: coming too late? Haematologica 2023; 108:661-662. [PMID: 35586968 PMCID: PMC9973469 DOI: 10.3324/haematol.2021.281255] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2022] [Indexed: 11/09/2022] Open
|
238
|
Alhaj Moustafa M. Timing of CAR-T therapy in follicular lymphoma. Expert Rev Anticancer Ther 2023; 23:347-349. [PMID: 36825538 DOI: 10.1080/14737140.2023.2185222] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/25/2023]
|
239
|
Vu K, Frank MJ. CAR T-cell therapy for mantle cell lymphoma with central nervous system relapse. Blood Adv 2023; 7:375-378. [PMID: 35895518 PMCID: PMC9898604 DOI: 10.1182/bloodadvances.2022008031] [Citation(s) in RCA: 9] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2022] [Revised: 07/01/2022] [Accepted: 07/17/2022] [Indexed: 02/01/2023] Open
Affiliation(s)
- Khoan Vu
- Department of Oncology/Hematology, Kaiser Permanente Santa Clara Medical Center, Santa Clara, CA
| | - Matthew J. Frank
- Division of Blood and Marrow Transplantation and Cellular Therapy, Stanford University, Palo Alto, CA
| |
Collapse
|
240
|
Fischer L, Dreyling M. Follicular lymphoma: an update on biology and optimal therapy. Leuk Lymphoma 2023; 64:761-775. [PMID: 37061956 DOI: 10.1080/10428194.2023.2174804] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/11/2023]
Abstract
Follicular lymphoma (FL) is a mature B-cell neoplasm with a mostly indolent course. Genetic hallmark characteristics are Bcl-2 overexpression based on a t(14;18) translocation and additional secondary genetic and epigenetic aberrations. Standard treatment for early-stage patients has curative intent and usually consists of radiotherapy with or without rituximab. In the advanced stage, the main therapeutic focus is on prolonged remissions. Therefore, treatment in asymptomatic patients is usually deferred. Symptomatic patients are subject to immunochemotherapy induction followed by antibody maintenance. Importantly, about one in five patients subsequently experiences a more rapid clinical course, achieving only short remissions with multiple relapses (POD24). In those patients, there is still an urgent need for improved therapeutic options. Accordingly, a plethora of targeted and immunotherapeutic options, including immunomodulatory drugs, small molecule inhibitors, monoclonal antibodies as well as bispecific T-cell engager antibodies and CAR-T cell products have been recently evaluated in such relapsed high-risk patients.
Collapse
Affiliation(s)
- Luca Fischer
- Department of Medicine III, University Hospital, LMU Munich, Munich, Germany
| | - Martin Dreyling
- Department of Medicine III, University Hospital, LMU Munich, Munich, Germany
| |
Collapse
|
241
|
Labanieh L, Mackall CL. CAR immune cells: design principles, resistance and the next generation. Nature 2023; 614:635-648. [PMID: 36813894 DOI: 10.1038/s41586-023-05707-3] [Citation(s) in RCA: 188] [Impact Index Per Article: 188.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2022] [Accepted: 01/04/2023] [Indexed: 02/24/2023]
Abstract
The remarkable clinical activity of chimeric antigen receptor (CAR) therapies in B cell and plasma cell malignancies has validated the use of this therapeutic class for liquid cancers, but resistance and limited access remain as barriers to broader application. Here we review the immunobiology and design principles of current prototype CARs and present emerging platforms that are anticipated to drive future clinical advances. The field is witnessing a rapid expansion of next-generation CAR immune cell technologies designed to enhance efficacy, safety and access. Substantial progress has been made in augmenting immune cell fitness, activating endogenous immunity, arming cells to resist suppression via the tumour microenvironment and developing approaches to modulate antigen density thresholds. Increasingly sophisticated multispecific, logic-gated and regulatable CARs display the potential to overcome resistance and increase safety. Early signs of progress with stealth, virus-free and in vivo gene delivery platforms provide potential paths for reduced costs and increased access of cell therapies in the future. The continuing clinical success of CAR T cells in liquid cancers is driving the development of increasingly sophisticated immune cell therapies that are poised to translate to treatments for solid cancers and non-malignant diseases in the coming years.
Collapse
Affiliation(s)
- Louai Labanieh
- Department of Bioengineering, Stanford University, Stanford, CA, USA.,Center for Cancer Cell Therapy, Stanford Cancer Institute, Stanford University, Stanford, CA, USA.,Parker Institute for Cancer Immunotherapy, San Francisco, CA, USA
| | - Crystal L Mackall
- Center for Cancer Cell Therapy, Stanford Cancer Institute, Stanford University, Stanford, CA, USA. .,Parker Institute for Cancer Immunotherapy, San Francisco, CA, USA. .,Division of Hematology, Oncology, Stem Cell Transplantation and Regenerative Medicine, Department of Pediatrics, Stanford University, Stanford, CA, USA. .,Division of Blood and Marrow Transplantation and Cell Therapy, Department of Medicine, Stanford University, Stanford, CA, USA.
| |
Collapse
|
242
|
Alhamhoom Y, Kakinani G, Rahamathulla M, Ali M. Osmani R, Hani U, Yoonus Thajudeen K, Kiran Raj G, Gowda DV. Recent advances in the liposomal nanovesicles based immunotherapy in the treatment of cancer: A review. Saudi Pharm J 2023; 31:279-294. [PMID: 36942270 PMCID: PMC10023551 DOI: 10.1016/j.jsps.2022.12.008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2022] [Accepted: 12/17/2022] [Indexed: 12/25/2022] Open
Abstract
Immunotherapy, along with chemotherapy, targeted delivery, radiation and surgery has become one of the most common cancer treatments. The aim of cancer immunology is to use the bodys immune system to combat tumors and develop a robust antitumor immune response. In the last few years, immune checkpoint inhibitors and chimeric antigen receptor-modified T cells have made substantial advancements in cancer immunotherapy. By boosting cell type-specific delivery and immunological responses, nanocarriers like liposomes have the ability to enhance greater immune responses. The efficacy of anti-tumor therapeutics is being significantly improved as liposomes can assist in resolving a number of issues that can arise from a variety of cancer immunotherapies. Since, liposomes can be loaded with both hydrophilic and hydrophobic drugs and protect the immunotherapeutic agents loaded inside the core, they offer significant advantages over other nano delivery systems. The use of liposomes for accurate and timely delivery of immunotherapies to particular targeted neoplasms, with little or no injury to healthy cells, maximizes immunotherapy efficacy. Liposomes are also suitable vehicles for delivering medications simultaneously with other therapies such as chemotherapy, radiation, and phototherapy. Liposomal nanoparticles will be introduced and used as an objective immunotherapy delivery system for great precision, making them a viable cancer treatment approach.With an emphasis on dendritic cells, T cells, tumor and natural killer cells, and macrophages; outline of many forms of immune-therapies in oncology and cutting-edge advances in liposomal nanovesicles for cancer immunotherapy are covered in this review.
Collapse
Affiliation(s)
- Yahya Alhamhoom
- Department of Pharmaceutics, College of Pharmacy, King Khalid University, Abha 62529, Saudi Arabia
| | - Greeshma Kakinani
- Department of Pharmaceutics, JSS College of Pharmacy, JSS Academy of Higher Education and Research, Mysuru, Karnataka, India
| | - Mohamed Rahamathulla
- Department of Pharmaceutics, College of Pharmacy, King Khalid University, Abha 62529, Saudi Arabia
| | - Riyaz Ali M. Osmani
- Department of Pharmaceutics, JSS College of Pharmacy, JSS Academy of Higher Education and Research, Mysuru, Karnataka, India
| | - Umme Hani
- Department of Pharmaceutics, College of Pharmacy, King Khalid University, Abha 62529, Saudi Arabia
| | - Kamal Yoonus Thajudeen
- Department of Pharmacognosy, College of Pharmacy, King Khalid University, Abha 62529, Saudi Arabia
| | - G. Kiran Raj
- Department of Pharmaceutics, JSS College of Pharmacy, JSS Academy of Higher Education and Research, Mysuru, Karnataka, India
| | - Devegowda V. Gowda
- Department of Pharmaceutics, Cauvery College of Pharmacy, Mysuru 570 028, Karnataka, India
| |
Collapse
|
243
|
Palomba ML, Ghione P, Patel AR, Nahas M, Beygi S, Hatswell AJ, Kanters S, Limbrick-Oldfield EH, Wade SW, Ray MD, Owen J, Neelapu SS, Gribben J, Radford J, Bobillo S. A 24-month updated analysis of the comparative effectiveness of ZUMA-5 (axi-cel) vs. SCHOLAR-5 external control in relapsed/refractory follicular lymphoma. Expert Rev Anticancer Ther 2023; 23:199-206. [PMID: 36723678 PMCID: PMC11104735 DOI: 10.1080/14737140.2023.2171994] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2022] [Accepted: 01/11/2023] [Indexed: 02/02/2023]
Abstract
BACKGROUND In the ZUMA-5 trial (Clinical trials identification: NCT03105336), axicabtagene ciloleucel (axi-cel; a chimeric antigen receptor T-cell therapy) demonstrated high rates of durable response in relapsed/refractory (r/r) follicular lymphoma (FL) patients and clear superiority relative to the SCHOLAR-5 external control cohort. We update this comparison using the ZUMA-5 24-month data. RESEARCH DESIGN AND METHODS The SCHOLAR-5 cohort is comprised of r/r FL patients who initiated ≥3rd line of therapy after July 2014 and meeting ZUMA-5 eligibility criteria. Groups were balanced for patient characteristics through propensity scoring on prespecified prognostic factors using standardized mortality ratio (SMR) weighting. The overall response rate was compared using a weighted logistic regression. Time-to-event outcomes were evaluated using a Cox regression. RESULTS For SCHOLAR-5, the sum of weights for the 143 patients was 85 after SMR weighting, versus 86 patients in ZUMA-5. The median follow-up was 29.4 months and 25.4 months for ZUMA-5 and SCHOLAR-5, respectively. The hazard ratios for overall survival and progression-free survival were 0.52 (95% confidence interval (CI): 0.28-0.95) and 0.28 (95% CI: 0.17-0.45), favoring axi-cel. CONCLUSION This updated analysis, using a longer minimum follow-up than a previously published analysis, shows that the improved efficacy of axi-cel, relative to available therapies, in r/r FL is durable. .
Collapse
Affiliation(s)
- M Lia Palomba
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Paola Ghione
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | | | - Myrna Nahas
- Kite, A Gilead Company, Santa Monica, CA, USA
| | - Sara Beygi
- Kite, A Gilead Company, Santa Monica, CA, USA
| | | | | | | | - Sally W Wade
- Wade Outcomes Research and Consulting, Salt Lake City, UT, USA
| | | | | | - Sattva S Neelapu
- The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | | | - John Radford
- The University of Manchester and Christie NHS Foundation Trust, Manchester Academic Health Science Centre, Manchester, UK
| | | |
Collapse
|
244
|
Smyth E, Cheah CY, Seymour JF. Management of indolent B-cell Lymphomas: A review of approved and emerging targeted therapies. Cancer Treat Rev 2023; 113:102510. [PMID: 36634434 DOI: 10.1016/j.ctrv.2023.102510] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2022] [Revised: 12/23/2022] [Accepted: 01/01/2023] [Indexed: 01/07/2023]
Abstract
The indolent B-cell non-Hodgkin lymphomas (B-NHL) comprise a heterogenous group of lymphoproliferative disorders characterized by slow growth kinetics and a relapsing/remitting course. Management has, until recently, been uniform across all indolent B-NHL subtypes. Improving insight into pathophysiological and molecular features of each disease has led to development of several targeted therapies. Consequently, each subtype must now be considered an individual entity. In this review, we consider the three commonest indolent B-NHLs: follicular lymphoma, marginal zone lymphoma and Waldenstrom's macroglobulinemia and review in detail the data on approved and emerging targeted therapeutic agents for each B-NHL subtype.
Collapse
Affiliation(s)
- Elizabeth Smyth
- Department of Hematology, Sir Charles Gairdner Hospital, Perth, Western Australia, Australia
| | - Chan Y Cheah
- Department of Hematology, Sir Charles Gairdner Hospital, Perth, Western Australia, Australia; Medical School, University of Western Australia, Perth, Western Australia, Australia.
| | - John F Seymour
- Peter MacCallum Cancer Centre & The Royal Melbourne Hospital, Melbourne, Victoria, Australia; University of Melbourne, Victoria, Australia.
| |
Collapse
|
245
|
Zhang X, Wu J, Qiao L, Chen L, Chen C, Zhang H, Luo R, Xiao Y. Case report: Cryoablation as a novel bridging strategy prior to CAR-T cell therapy for B cell malignancies with bulky disease. Front Oncol 2023; 13:1008828. [PMID: 36776338 PMCID: PMC9911860 DOI: 10.3389/fonc.2023.1008828] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2022] [Accepted: 01/17/2023] [Indexed: 01/28/2023] Open
Abstract
Chimeric antigen receptor (CAR) T-cell therapy has emerged as a powerful immunotherapy in relapsed/refractory (R/R) hematological malignancies, especially in R/R B-cell acute lymphocytic leukemia (B-ALL), non-Hodgkin lymphoma (NHL), and multiple myeloma (MM). To prevent disease progression and reduce tumor burden during CAR-T cell manufacturing, bridging therapies prior to CAR-T cell infusion are crucial. At present, it has been demonstrated that targeted therapy, radiotherapy and autologous stem cell transplantation (ASCT) could serve as effective bridging strategies. However, whether cryoablation could serve as a novel bridging strategy is unknown. In this paper, we report 2 cases of R/R B cell malignancies with bulky disease that were successfully treated with a combination of cryoablation and CAR-T cell therapy. Patient 1 was a 65-year-old female who was diagnosed with R/R MM with extramedullary disease (EMD). She was enrolled in the anti-BCMA CAR-T cell clinical trial. Patient 2 was a 70-year-old man who presented with a subcutaneous mass in the right anterior thigh and was diagnosed with primary cutaneous diffuse large B cell lymphoma, leg type (PCLBCL-LT) 1 year ago. He failed multiline chemotherapies as well as radiotherapy. Thus, he requested anti-CD19 CAR-T cell therapy. Unfortunately, they all experienced local progression during CAR-T cell manufacturing. To rapidly achieve local tumor control and reduce tumor burden, they both received cryoablation as a bridging therapy. Patient 1 achieved a very good partial response (VGPR) 1 month after CAR-T cell infusion, and patient 2 achieved a partial response (PR) 1 month after CAR-T cell infusion. In addition, adverse effects were tolerable and manageable. Our study demonstrated the favorable safety and efficacy of combination therapy with cryoablation and CAR-T cell therapy for the first time, and it also indicates that cryoablation could serve as a novel therapeutic strategy for local tumor control in B cell malignancies.
Collapse
Affiliation(s)
- Xiaomin Zhang
- Department of Hematology, Jinshazhou Hospital of Guangzhou University of Chinese Medicine, Guangzhou, China,Institute of Clinical Medicine College, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Jinming Wu
- Department of Hematology, Jinshazhou Hospital of Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Liangliang Qiao
- Department of Hematology, Jinshazhou Hospital of Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Lixuan Chen
- Department of Hematology, Jinshazhou Hospital of Guangzhou University of Chinese Medicine, Guangzhou, China,Institute of Clinical Medicine College, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Chaolin Chen
- Translational Medicine Center, The Second Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Hui Zhang
- School of Medicine, Jishou University, Jishou, China
| | - Rongcheng Luo
- Thoracic Surgery, Jinshazhou Hospital of Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Yang Xiao
- Institute of Clinical Medicine College, Guangzhou University of Chinese Medicine, Guangzhou, China,Department of Hematology, Shenzhen Qianhai Shekou Pilot Free Trade Zone Hospital, Shenzhen, China,*Correspondence: Yang Xiao,
| |
Collapse
|
246
|
Kanters S, Ball G, Kahl B, Wiesinger A, Limbrick-Oldfield EH, Sudhindra A, Snider JT, Patel AR. Clinical outcomes in patients relapsed/refractory after ≥2 prior lines of therapy for follicular lymphoma: a systematic literature review and meta-analysis. BMC Cancer 2023; 23:74. [PMID: 36690960 PMCID: PMC9869623 DOI: 10.1186/s12885-023-10546-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2022] [Accepted: 01/13/2023] [Indexed: 01/25/2023] Open
Abstract
BACKGROUND Patients with follicular lymphoma (FL) can have high response rates to early lines of treatment. However, among FL patients relapsed/refractory (r/r) after ≥2 prior lines of therapy (LOT), remission tends to be shorter and there is limited treatment guidance. This study sought to evaluate the clinical outcomes for r/r FL after ≥2 prior LOT identified through systematic literature review. METHODS Eligible studies included comparative or non-comparative interventional or observational studies of systemic therapies among adults with FL r/r after ≥2 prior LOT published prior to 31st May 2021. Prior LOT must have included an anti-CD20 monoclonal antibody and an alkylating agent, in combination or separately. Overall response rate (ORR) and complete response (CR) were estimated using inverse-variance weighting with Freeman-Tukey double-arcsine transformations. Kaplan-Meier (KM) curves for progression-free survival (PFS) and overall survival (OS) estimated by reconstructing digitized curves using the Guyot algorithm, and survival analyses were conducted, stratified by ≥2 prior LOT and ≥ 3 prior LOT groups (as defined in the source material). Restricting the analyses to the observational cohorts was investigated as a sensitivity analysis. RESULTS The analysis-set included 20 studies published between 2014 and 2021. Studies were primarily US and/or European based, with the few exceptions using treatments approved in US/Europe. The estimated ORR was 58.47% (95% confidence interval [CI]: 51.13-65.62) and proportion of patients with CR was 19.63% (95% CI: 15.02-24.68). The median OS among those ≥2 prior LOT was 56.57 months (95% CI: 47.8-68.78) and median PFS was 9.78 months (95% CI: 9.01-10.63). The 24-month OS decreased from 66.50% in the ≥2 prior LOT group to 59.51% in the ≥3 prior LOT group, with a similar trend in PFS at 24-month (28.42% vs 24.13%). CONCLUSIONS This study found that few r/r FL patients with ≥2 prior LOT achieve CR, and despite some benefit, approximately 1/3 of treated patients die within 24 months. The shorter median PFS with increasing prior LOT suggest treatment durability is suboptimal in later LOT. These findings indicate that patients are underserved by treatments currently available in the US and Europe.
Collapse
Affiliation(s)
| | - Graeme Ball
- grid.437263.7Gilead Sciences Canada, Inc., Mississauga, Canada
| | - Brad Kahl
- grid.4367.60000 0001 2355 7002Oncology Division, Department of Medicine, Washington University School of Medicine in St. Louis, St. Louis, MO USA
| | | | | | | | | | - Anik R. Patel
- grid.504964.aKite, A Gilead Company, Santa Monica, USA
| |
Collapse
|
247
|
Câmara AB, Brandão IA. The Non-Hodgkin Lymphoma Treatment and Side Effects: A Systematic Review and Meta-Analysis. Recent Pat Anticancer Drug Discov 2023; 19:PRA-EPUB-128894. [PMID: 36650656 DOI: 10.2174/1574892818666230117151757] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2022] [Revised: 10/11/2022] [Accepted: 11/11/2022] [Indexed: 01/19/2023]
Abstract
OBJECTIVE This paper aims to review studies regarding side effects found during Non-Hodgkin Lymphoma treatment, to suggest the drug class most associated with these effects, as well as the most prevalent side effect grade. METHODS This review is registered in PROSPERO (IDCRD42022295774) and followed the PICOS strategy and PRISMA guidelines. The search was carried out in the databases PubMed/MEDLINE, Scientific Electronic Library Online, and DOAJ. Medical Subject Headings Terms were used and quantitative studies with conclusive results regarding side effects during the non-Hodgkin lymphoma treatment were selected. Patent information was obtained from google patents. RESULTS Monoclonal antibodies were the main drug class associated with side effects during NHL therapy. The combination of Rituximab (Rituxan®; patent EP1616572B) and iInotuzumab (Besponsa®; patent EP1504035B3) was associated with a higher incidence of thrombocytopenia (p<0.05), while the combination of Rituximab and Venetoclax (Venclexta®; patent CN107089981A) was associated with a higher incidence of neutropenia (p<0.05) when compared to Bendamustine combinations (Treanda ™; patent US20130253025A1). Meta-analysis revealed a high prevalence of grade 3-4 neutropenia and thrombocytopenia in men. Finally, Americans and Canadians experienced a higher prevalence of these side effects, when compared to others nationalities (p<0.05). CONCLUSION Patents regarding the use of monoclonal antibodies in NHL treatment were published in the last year. Monoclonal antibodies associated with neutropenia (grade 3-4) and thrombocytopenia, especially in North American men treated for NHL, and with an average age of 62 years demonstrated importance in this study.
Collapse
Affiliation(s)
- Alice Barros Câmara
- Department of Biophysics and Pharmacology, Federal University of Rio Grande do Norte
| | - Igor Augusto Brandão
- Bioinformatics Multidisciplinary Environment, Federal University of Rio Grande do Norte
| |
Collapse
|
248
|
Puckrin R, Chua N, Chin K, Peters A, Duggan P, Shafey M, Storek J, Jamani K, Owen C, Stewart D. Long-term follow-up demonstrates curative potential of autologous stem cell transplantation for relapsed follicular lymphoma. Br J Haematol 2023; 201:319-325. [PMID: 36625160 DOI: 10.1111/bjh.18640] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2022] [Revised: 12/16/2022] [Accepted: 12/22/2022] [Indexed: 01/11/2023]
Abstract
Although autologous stem cell transplantation (ASCT) can achieve durable responses in eligible patients with follicular lymphoma (FL), long-term follow-up is needed to determine if it has curative potential. This retrospective, multicenter study included 162 patients who received ASCT for relapsed FL in Alberta, Canada. With a median (range) follow-up time of 12.5 years (0.1-27.9), the 12-year time-to-progression (TTP) was 57% (95% confidence interval [CI] 49%-65%), time-to-next-treatment was 61% (95% CI 52%-69%), progression-free survival was 51% (95% CI 42%-59%) and overall survival was 69% (95% CI 60%-76%). A plateau emerged on the TTP curve at 57% starting 9 years after ASCT with no relapses occurring beyond this timepoint. Ten patients remained in remission 20 years or more after ASCT. Patients undergoing ASCT at first or second relapse had superior outcomes compared to third or later relapse (12-year TTP 61% vs. 34%), as did patients without progression of disease within 24 months (POD24) of frontline treatment versus those with POD24 (12-year TTP 67% vs. 50%). ASCT achieves high rates of durable remission in relapsed FL, with long-term follow-up revealing that more than 50% of transplanted patients may be functionally cured of their lymphoma. The optimal timing to consider ASCT is at first or second relapse, regardless of POD24 status.
Collapse
Affiliation(s)
- Robert Puckrin
- Tom Baker Cancer Centre and University of Calgary, Calgary, Canada
| | - Neil Chua
- Cross Cancer Institute and University of Alberta, Edmonton, Canada
| | - Kelly Chin
- Tom Baker Cancer Centre and University of Calgary, Calgary, Canada
| | - Anthea Peters
- Cross Cancer Institute and University of Alberta, Edmonton, Canada
| | - Peter Duggan
- Tom Baker Cancer Centre and University of Calgary, Calgary, Canada
| | - Mona Shafey
- Tom Baker Cancer Centre and University of Calgary, Calgary, Canada
| | - Jan Storek
- Tom Baker Cancer Centre and University of Calgary, Calgary, Canada
| | - Kareem Jamani
- Tom Baker Cancer Centre and University of Calgary, Calgary, Canada
| | - Carolyn Owen
- Tom Baker Cancer Centre and University of Calgary, Calgary, Canada
| | - Douglas Stewart
- Tom Baker Cancer Centre and University of Calgary, Calgary, Canada
| |
Collapse
|
249
|
Epperla N, Zhao Q, Chowdhury SM, Shea L, Moyo TK, Reddy N, Sheets J, Weiner DM, Geethakumari PR, Kandarpa M, Bruno XJ, Thomas C, Churnetski MC, Hsu A, Zurbriggen L, Tan XWC, Lindsey K, Maakaron J, Caimi PF, Torka P, Bello C, Ayyappan S, Oh TS, Karmali R, Kim SH, Kress A, Kothari S, Sawalha Y, Christian B, David KA, Greenwell IB, Janakiram M, Kenkre VP, Olszewski AJ, Cohen JB, Palmisiano N, Umyarova E, Wilcox RA, Awan FT, Alderuccio JP, Barta SK, Grover NS, Ghosh N, Bartlett NL, Herrera AF, Shouse G. Postibrutinib relapse outcomes for patients with marginal zone lymphoma. Blood Adv 2023; 7:88-91. [PMID: 36269847 PMCID: PMC9827027 DOI: 10.1182/bloodadvances.2022008634] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2022] [Revised: 09/07/2022] [Accepted: 09/24/2022] [Indexed: 01/18/2023] Open
Affiliation(s)
- Narendranath Epperla
- Division of Hematology, The Ohio State University, Columbus, OH
- Correspondence: Narendranath Epperla, Division of Hematology, Department of Medicine, The Ohio State University, Columbus, OH 43210;
| | - Qiuhong Zhao
- Division of Hematology, The Ohio State University, Columbus, OH
| | | | - Lauren Shea
- Division of Hematology, Washington University, St. Louis, MO
| | | | | | - Julia Sheets
- Division of Hematology and Oncology, University of North Carolina, Chapel Hill, NC
| | - David M. Weiner
- Division of Hematology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA
| | | | - Malathi Kandarpa
- Division of Hematology, Rogel Cancer Center, University of Michigan, Ann Arbor, MI
| | | | - Colin Thomas
- Division of Hematology, Thomas Jefferson University, Philadelphia, PA
| | | | - Andrew Hsu
- Division of Hematology, Brown University, Providence, RI
| | - Luke Zurbriggen
- Division of Hematology, University of Wisconsin, Madison, WI
| | | | - Kathryn Lindsey
- Division of Hematology and Oncology, Medical University of South Carolina, Charleston, SC
| | - Joseph Maakaron
- Division of Hematology, University of Minnesota, Minneapolis, MN
| | - Paolo F. Caimi
- University Hospitals Seidman Cancer Center, Cleveland, OH
| | - Pallawi Torka
- Department of Medicine, Roswell Park Cancer Institute, Buffalo, NY
| | - Celeste Bello
- Division of Hematology, H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL
| | - Sabarish Ayyappan
- Division of Hematology and Oncology, University of Iowa, Iowa City, IA
| | - Timothy S. Oh
- Division of Hematology, Northwestern University, Chicago, IL
| | - Reem Karmali
- Division of Hematology, Northwestern University, Chicago, IL
| | - Seo-Hyun Kim
- Division of Hematology and Oncology, Rush University, Chicago, IL
| | - Anna Kress
- Division of Hematology, Yale University, New Haven, CT
| | | | - Yazeed Sawalha
- Division of Hematology, The Ohio State University, Columbus, OH
| | - Beth Christian
- Division of Hematology, The Ohio State University, Columbus, OH
| | | | - Irl Brian Greenwell
- Division of Hematology and Oncology, Medical University of South Carolina, Charleston, SC
| | - Murali Janakiram
- Division of Hematology, University of Minnesota, Minneapolis, MN
| | | | | | - Jonathon B. Cohen
- Winship Cancer Institute, Emory University Medical Center, Atlanta, GA
| | - Neil Palmisiano
- Division of Hematology, Thomas Jefferson University, Philadelphia, PA
| | - Elvira Umyarova
- Division of Hematology and Oncology, University of Vermont, Burlington, VT
| | - Ryan A. Wilcox
- Division of Hematology, Rogel Cancer Center, University of Michigan, Ann Arbor, MI
| | - Farrukh T. Awan
- Harold C. Simmons Comprehensive Cancer Center, University of Texas Southwestern Medical Center, Dallas, TX
| | | | - Stefan K. Barta
- Division of Hematology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA
| | - Natalie S. Grover
- Division of Hematology and Oncology, University of North Carolina, Chapel Hill, NC
| | | | | | | | | |
Collapse
|
250
|
Davis JA, Gaffney KJ, McGann M, Smith D, Edwards K, Baldino E, Bakos K, Butcher C, Greenwell B, Hess BT, Hashmi H. Fever Characteristics and Impact on Safety and Efficacy of Chimeric Antigen Receptor T-Cell Therapy. CLINICAL LYMPHOMA, MYELOMA & LEUKEMIA 2023; 23:e14-e18. [PMID: 36319568 DOI: 10.1016/j.clml.2022.09.005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/21/2022] [Revised: 09/29/2022] [Accepted: 09/30/2022] [Indexed: 11/08/2022]
Abstract
BACKGROUND Fever is a hallmark symptom of cytokine release syndrome (CRS) after chimeric antigen receptor (CAR) T-cell therapy. Fever characteristics and the impact of fever on safety and efficacy post CAR T are not well understood. We sought to explore the impact of fever and its characteristics on safety and efficacy post CAR T-cell therapy. PATIENTS AND METHODS We reviewed 40 patients with various hematologic malignancies (non-Hodgkin lymphoma, acute lymphoblastic leukemia, multiple myeloma) treated with CAR T-cell therapy between March 2019 and March 2022. We evaluated all patients who developed fever after CAR T infusion and analyzed the association of fever with toxicity (CRS and neurotoxicity) and efficacy (overall response (ORR) and complete response (CR) at day +90 post CAR T infusion). Fever was defined as per Lee criteria (equal to or greater than 38°C). CRS and immune-effector cell associated neurotoxicity syndrome (ICANS) were graded using American Society for Transplantation and Cellular Therapy grading system. RESULTS Fever occurred in 75% (30/40) of patients. Rates of all grade and grade 3+ CRS and ICANS were 75%, 2%, 33% and 10%, respectively. Fever occurred within 24 and 72 hours after CAR T infusion in 40% and 53% of patients, respectively. Fifty percent of patients received tocilizumab (toci) for CRS. After the first dose of toci, fever recurred in 38% of the patients, of which 67% had recurrence within 24 hours. Day +90 CR rates were 43% and 10% in patients with and without fever, respectively (Table 3). CONCLUSION While fever is common after CAR T-cell therapy, early-onset and higher magnitude do not appear to affect safety or efficacy of CAR T. Absence of fever may affect response to CAR T.
Collapse
Affiliation(s)
- James A Davis
- Department of Malignant Hematology and Bone Marrow Transplant, Medical University of South Carolina, Charleston, SC.
| | - Kelly J Gaffney
- Department of Malignant Hematology and Bone Marrow Transplant, Medical University of South Carolina, Charleston, SC
| | - Mary McGann
- Department of Malignant Hematology and Bone Marrow Transplant, Medical University of South Carolina, Charleston, SC
| | - Deidra Smith
- Department of Malignant Hematology and Bone Marrow Transplant, Medical University of South Carolina, Charleston, SC
| | - Kathy Edwards
- Department of Malignant Hematology and Bone Marrow Transplant, Medical University of South Carolina, Charleston, SC
| | - Ellen Baldino
- Department of Malignant Hematology and Bone Marrow Transplant, Medical University of South Carolina, Charleston, SC
| | - Keegan Bakos
- Department of Malignant Hematology and Bone Marrow Transplant, Medical University of South Carolina, Charleston, SC
| | - Coleen Butcher
- Department of Malignant Hematology and Bone Marrow Transplant, Medical University of South Carolina, Charleston, SC
| | - Brian Greenwell
- Department of Malignant Hematology and Bone Marrow Transplant, Medical University of South Carolina, Charleston, SC
| | - Brian T Hess
- Department of Malignant Hematology and Bone Marrow Transplant, Medical University of South Carolina, Charleston, SC
| | - Hamza Hashmi
- Department of Malignant Hematology and Bone Marrow Transplant, Medical University of South Carolina, Charleston, SC
| |
Collapse
|