1
|
Mahmoudjafari Z, Li J, Bercaw E, Parisé H, Bognar K, Wang ST, Masaquel A. Budget impact of introducing glofitamab for treatment of relapsed or refractory diffuse large B-cell lymphoma after two or more lines of systemic therapy in the United States. J Med Econ 2025; 28:595-604. [PMID: 40163049 DOI: 10.1080/13696998.2025.2486839] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/17/2024] [Accepted: 03/27/2025] [Indexed: 04/02/2025]
Abstract
BACKGROUND Glofitamab is a T-cell engaging bispecific monoclonal antibody that was granted accelerated approval from the United States Food and Drug Administration for adult patients with relapsed or refractory diffuse large B-cell lymphoma (DLBCL), not otherwise specified or large B-cell lymphoma arising from follicular lymphoma, after ≥2 lines of systemic therapy (3L+). METHODS A budget impact model was developed for a hypothetical blended commercial/Medicare health plan with 1,000,000 members. Comparators were axicabtagene ciloleucel (Axi-cel), lisocabtagene maraleucel (Liso-cel), tisagenlecleucel (Tisa-cel), loncastuximab tesirine, polatuzumab vedotin + bendamustine + rituximab, rituximab + gemcitabine + oxaliplatin, tafasitamab + lenalidomide, and epcoritamab (Epcor). Total costs included those for drugs, wastage, administration, grade ≥3 adverse reactions, and all-grade cytokine release syndrome) and routine care. Market shares were based on internal projections and expert opinions. Total and per-member per-month (PMPM) net budget impacts over 3 years were calculated. RESULTS Approximately nine patients were projected to be eligible for 3L + DLBCL treatment in a health plan of 1,000,000 members. The introduction of glofitamab as a treatment option resulted in estimated total and PMPM cost savings of $728,697 and -$0.0202, respectively, over 3 years. Costs were reduced across all cost categories but particularly in drug costs. Among the newer therapies, total 3-year cost per treated patient was lowest for glofitamab: $226,658 versus Tisa-cel = $564,113; Axi-cel = $540,002; Liso-cel = $516,272; and Epcor = $335,293. Across all sensitivity analyses, the inclusion of glofitamab had minimal PMPM budget impact, ranging from -$0.0256 to -$0.0108. CONCLUSIONS With the lowest 3-year total cost per treated patient among the newer therapies, glofitamab being an available option in the 3L + DLBCL market is estimated to save a hypothetical 1,000,000-member health plan $728,697 in cumulative total costs and $0.0202 in PMPM costs over 3 years.
Collapse
Affiliation(s)
| | - Jia Li
- Genentech, Inc, South San Francisco, CA, USA
| | | | | | | | | | | |
Collapse
|
2
|
Locke FL, Siddiqi T, Jacobson CA, Nikiforow S, Ahmed S, Miklos DB, Lin Y, Lunning MA, Hill BT, Ghobadi A, Hu ZH, Hemmer MT, Zoratti MJ, Vunnum S, Tsang J, Spooner C, Smith H, Fu C, Patel A, Miao H, Shahani SA, Mirjah DL, Xu H, Pasquini MC. Impact of vein-to-vein time in patients with R/R LBCL treated with axicabtagene ciloleucel. Blood Adv 2025; 9:2663-2676. [PMID: 39883946 DOI: 10.1182/bloodadvances.2024013656] [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: 07/25/2024] [Revised: 11/26/2024] [Accepted: 12/30/2024] [Indexed: 02/01/2025] Open
Abstract
ABSTRACT Chimeric antigen receptor (CAR) T-cell products axicabtagene ciloleucel (axi-cel), tisagenlecleucel (tisa-cel), and lisocabtagene maraleucel (liso-cel) are approved for relapsed/refractory (R/R) large B-cell lymphoma (LBCL). Emerging evidence indicates that delayed CAR T-cell infusion, including prolonged time from leukapheresis to infusion, known as vein-to-vein time (V2Vt), may adversely impact clinical outcomes. We conducted a systematic literature review (SLR) and meta-analysis to identify differences in V2Vt in patients with R/R LBCL treated with axi-cel, tisa-cel, or liso-cel. The impact of V2Vt (<28 days vs ≥28 to <40 days vs ≥40 days) on effectiveness and safety outcomes was evaluated in patients treated with axi-cel enrolled in a post-authorization safety study using the Center for International Blood and Marrow Transplant Research data. SLR and meta-analysis showed that patients treated with axi-cel had the shortest median V2Vt (30.6 days) compared with tisa-cel (48.4 days) or liso-cel (35.9 days). Real-world analysis of patients treated with axi-cel demonstrated that V2Vt ≥40 days was associated with significantly lower complete response rate than V2Vt <28 days (odds ratio [OR], 0.61) or ≥28 to <40 days (OR, 0.66) and significantly worse overall survival than V2Vt <28 days (hazard ratio [HR], 1.33) or ≥28 to <40 days (HR, 1.36). Higher prolonged thrombocytopenia rates were observed in patients with axi-cel V2Vt ≥28 to <40 days or ≥40 days compared with <28 days (OR, 1.44 or 1.95, respectively). Together, these results show the impact of V2Vt on patient outcomes with axi-cel therapy and that earlier infusion with CD19-CAR therapies may be beneficial.
Collapse
MESH Headings
- Humans
- Lymphoma, Large B-Cell, Diffuse/therapy
- Lymphoma, Large B-Cell, Diffuse/mortality
- Lymphoma, Large B-Cell, Diffuse/drug therapy
- Immunotherapy, Adoptive/methods
- Immunotherapy, Adoptive/adverse effects
- Antigens, CD19/therapeutic use
- Treatment Outcome
- Veins
- Time Factors
- Receptors, Chimeric Antigen
- Biological Products
- Receptors, Antigen, T-Cell
Collapse
Affiliation(s)
- Frederick L Locke
- Department of Blood and Marrow Transplant and Cellular Immunotherapy, Moffit Cancer Center, Tampa, FL
| | - Tanya Siddiqi
- Department of Hematology/HCT, City of Hope National Medical Center, Duarte, CA
| | - Caron A Jacobson
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA
| | - Sarah Nikiforow
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA
| | - Sairah Ahmed
- Department of Lymphoma - Myeloma, MD Anderson Cancer Center, Houston, TX
| | - David B Miklos
- Department of Medicine - Med/Blood and Marrow Transplantation, Stanford University School of Medicine, Stanford, CA
| | - Yi Lin
- Division of Hematology, Mayo Clinic, Rochester, MN
| | - Matthew A Lunning
- Division of Oncology and Hematology, Department of Internal Medicine, Fred & Pamela Buffett Cancer Center, Omaha, NE
| | - Brian T Hill
- Department of Hematology and Medical Oncology, Cleveland Clinic Taussig Cancer Institute, Cleveland, OH
| | - Armin Ghobadi
- Division of Oncology, Washington University School of Medicine, St. Louis, MO
| | | | | | | | | | | | | | | | | | - Anik Patel
- Kite, a Gilead Company, Santa Monica, CA
| | - Harry Miao
- Kite, a Gilead Company, Santa Monica, CA
| | | | | | - Hairong Xu
- Kite, a Gilead Company, Santa Monica, CA
| | - Marcelo C Pasquini
- Department of Hematology and Oncology - Medicine, Center for International Blood and Marrow Transplant Research, Milwaukee, WI
| |
Collapse
|
3
|
Io K, Nagai K, Kakihara B, Izumi K, Kitagawa T. Efficacy and safety of the thiotepa-busulfan conditioning regimen as for autologous stem cell transplantation in relapsed/refractory systemic diffuse large B cell lymphoma: a single-center retrospective study. Int J Hematol 2025; 121:813-819. [PMID: 39937333 DOI: 10.1007/s12185-025-03946-w] [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: 08/31/2024] [Revised: 01/29/2025] [Accepted: 01/29/2025] [Indexed: 02/13/2025]
Abstract
High-dose chemotherapy followed by autologous stem cell transplantation (ASCT) is a standard treatment for relapsed diffuse large B cell lymphoma (DLBCL). The BEAM regimen is widely used, but the MEAM regimen is more common in Japan due to the unavailability of carmustine. This retrospective analysis evaluated the efficacy of the thiotepa and busulfan (TT/BU) regimen compared with other regimens in 27 patients with systemic DLBCL who underwent ASCT at our institution from December 2013 to March 2022. Fourteen patients received the TT/BU regimen, while 13 received alternative regimens. The TT/BU regimen demonstrated superior progression-free survival (PFS) and overall survival (OS) compared to other regimens, with a 3 year PFS of 84.4% and OS of 91.7%. The TT/BU group also had fewer severe adverse events, particularly regarding renal function. Our findings suggest that the TT/BU regimen is a well-tolerated and effective alternative for relapsed/refractory DLBCL and provide valuable insights for future treatment strategies.
Collapse
Affiliation(s)
- Katsuhiro Io
- Department of Hematology, Kansai Electric Power Hospital, 2-1-7 Fukushima, Fukushima-ku, Osaka, 553-0003, Japan.
| | - Kenichi Nagai
- Department of Hematology, Kansai Electric Power Hospital, 2-1-7 Fukushima, Fukushima-ku, Osaka, 553-0003, Japan
| | - Bunta Kakihara
- Department of Hematology, Kansai Electric Power Hospital, 2-1-7 Fukushima, Fukushima-ku, Osaka, 553-0003, Japan
| | - Kiyotaka Izumi
- Department of Hematology, Kansai Electric Power Hospital, 2-1-7 Fukushima, Fukushima-ku, Osaka, 553-0003, Japan
| | - Tomoya Kitagawa
- Department of Hematology, Kansai Electric Power Hospital, 2-1-7 Fukushima, Fukushima-ku, Osaka, 553-0003, Japan
| |
Collapse
|
4
|
Gambella M, Carlomagno S, Raiola AM, Sivori S, Angelucci E. (CAR-)T cell dynamics following chimeric antigen receptor T cells for large B cell lymphoma: a translational tale. Leuk Lymphoma 2025; 66:1036-1044. [PMID: 39945648 DOI: 10.1080/10428194.2025.2456096] [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: 10/25/2024] [Revised: 12/17/2024] [Accepted: 01/15/2025] [Indexed: 05/27/2025]
Abstract
Chimeric antigen receptor (CAR) T-cell therapy represents a breakthrough in the treatment of B-cell malignancies. CAR-T cells infusion generally follows a chemotherapy regimen whose lymphodepleting properties create a favorable environment for the expansion of engineered T cells. While this process appears straightforward, emerging evidence reveals that complex mechanisms, collectively representing immune dynamics following CAR-T cell infusion, influence CAR-T cells behavior. In advance of infusion, a final-product enriched with less stressed CAR-T cells can improve their expansion and persistence, providing a biological rationale for early apheresis and administration. Following infusion, the emergence of dysfunctional CAR-T subpopulations, like regulatory or NK-like CAR-T cells, can impair efficacy. The recovery of non-CAR transduced T cells adds further complexity, as these cells could either impact outcomes or exacerbate complications, such as infections or prolonged cytopenia. In this review, we summarize the latest advances in understanding the immune dynamics following CAR-T cell infusion for large B-cell lymphomas, with a focus on both CAR-engineered and native T cell populations, and their impact on treatment efficacy and patient outcomes.
Collapse
MESH Headings
- Humans
- Receptors, Chimeric Antigen/metabolism
- Receptors, Chimeric Antigen/immunology
- Receptors, Chimeric Antigen/genetics
- Immunotherapy, Adoptive/methods
- Immunotherapy, Adoptive/adverse effects
- Lymphoma, Large B-Cell, Diffuse/therapy
- Lymphoma, Large B-Cell, Diffuse/immunology
- Lymphoma, Large B-Cell, Diffuse/pathology
- T-Lymphocytes/immunology
- T-Lymphocytes/metabolism
- Animals
- Receptors, Antigen, T-Cell/metabolism
Collapse
Affiliation(s)
- M Gambella
- IRCCS Ospedale Policlinico San Martino, Genova, Italy
- Department of Experimental Medicine (DIMES), University of Genoa, Genova, Italy
| | - S Carlomagno
- Department of Medicine (DMED), University of Udine, Udine, Italy
| | - A M Raiola
- IRCCS Ospedale Policlinico San Martino, Genova, Italy
| | - S Sivori
- IRCCS Ospedale Policlinico San Martino, Genova, Italy
- Department of Experimental Medicine (DIMES), University of Genoa, Genova, Italy
| | - E Angelucci
- IRCCS Ospedale Policlinico San Martino, Genova, Italy
| |
Collapse
|
5
|
Chen DT, Goloubeva O, Rapoport AP, Dahiya S, Atanackovic D, Hardy N, Kocoglu M, Lutfi F, Alkhaldi H, Claiborne JP, Lee ST, Kline K, Law JY, Yared JA. CD19 CAR-T With Axicabtagene Ciloleucel in R/R Large B-Cell Lymphoma With/Without Prior Autologous Stem Cell Transplant. CLINICAL LYMPHOMA, MYELOMA & LEUKEMIA 2025; 25:432-439. [PMID: 39865000 DOI: 10.1016/j.clml.2024.12.019] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/15/2024] [Revised: 12/24/2024] [Accepted: 12/28/2024] [Indexed: 01/28/2025]
Abstract
BACKGROUND Anti-CD19 CAR-T therapy has been a breakthrough in treatment of primary refractory or relapsed large B-cell lymphoma (r/r LBCL) and is poised to supplant previous second line of high dose chemotherapy and autologous stem cell transplantation (HDT/ASCT). However, in clinical practice, high risk patients with chemoimmunotherapy sensitive disease continue to receive salvage chemoimmunotherapy or cannot access CAR-T in a timely manner and thus may still proceed to HDT/ASCT. Little is known about clinical outcomes of CAR-T in patients who receive HDT/ASCT compared to those who are transplant-naïve. DESIGN We conducted a retrospective study of patients with r/r LBCL who previously underwent HDT/ASCT or were transplant-naïve (n = 97) and received axicabtagene ciloleucel after at least 2 prior therapy lines between 1/1/2018 to 12/31/2021. Primary endpoint was progression-free survival (PFS). Secondary endpoints were overall survival (OS), nonrelapse mortality (NRM), and cumulative incidence of relapse/progression. RESULTS 82 (84.5%) patients were transplant-naïve and 15 (15.5%) previously received HDT/ASCT. No differences were found in the incidence of high-grade cytokine release syndrome or immune effector cell-associated neurotoxicity syndrome, length of hospital admission, or incidence of cytopenia at day 30. 90-day response, PFS, OS, cumulative incidence of relapse/progression, and NRM were not different. Factors that adversely affected outcomes were prior bridging therapy, elevated LDH or thrombocytopenia at time of lymphodepleting chemotherapy, and worse ECOG performance status. CONCLUSION Prior treatment with HDT/ASCT does not compromise the safety and efficacy of anti-CD19 CAR-T therapy, suggesting a continued role for HDT/ASCT in treatment of select patients with r/r DLBCL.
Collapse
MESH Headings
- Humans
- Male
- Female
- Middle Aged
- Lymphoma, Large B-Cell, Diffuse/therapy
- Lymphoma, Large B-Cell, Diffuse/mortality
- Lymphoma, Large B-Cell, Diffuse/drug therapy
- Immunotherapy, Adoptive/methods
- Retrospective Studies
- Antigens, CD19/immunology
- Antigens, CD19/therapeutic use
- Transplantation, Autologous/methods
- Aged
- Adult
- Hematopoietic Stem Cell Transplantation/methods
- Biological Products/therapeutic use
- Biological Products/pharmacology
Collapse
Affiliation(s)
- David T Chen
- Division of Hematology/Oncology, Department of Medicine, Greenebaum Comprehensive Cancer Center, University of Maryland Medical Center, University of Maryland School of Medicine, Baltimore, MD.
| | - Olga Goloubeva
- Department of Epidemiology and Public Health, Division of Biostatistics and Bioinformatics, University of Maryland Marlene and Stewart Greenebaum Comprehensive Cancer Center, University of Maryland School of Medicine, Baltimore, MD
| | - Aaron P Rapoport
- Transplantation & Cellular Therapy Program, Division of Hematology/Oncology, Department of Internal Medicine, Greenebaum Comprehensive Cancer Center, University of Maryland Medical Center, University of Maryland School of Medicine, Baltimore, MD
| | - Saurabh Dahiya
- Transplantation & Cellular Therapy Program, Division of Hematology/Oncology, Department of Internal Medicine, Greenebaum Comprehensive Cancer Center, University of Maryland Medical Center, University of Maryland School of Medicine, Baltimore, MD
| | - Djordje Atanackovic
- Transplantation & Cellular Therapy Program, Division of Hematology/Oncology, Department of Internal Medicine, Greenebaum Comprehensive Cancer Center, University of Maryland Medical Center, University of Maryland School of Medicine, Baltimore, MD
| | - Nancy Hardy
- Transplantation & Cellular Therapy Program, Division of Hematology/Oncology, Department of Internal Medicine, Greenebaum Comprehensive Cancer Center, University of Maryland Medical Center, University of Maryland School of Medicine, Baltimore, MD
| | - Mehmet Kocoglu
- Transplantation & Cellular Therapy Program, Division of Hematology/Oncology, Department of Internal Medicine, Greenebaum Comprehensive Cancer Center, University of Maryland Medical Center, University of Maryland School of Medicine, Baltimore, MD
| | - Forat Lutfi
- Transplantation & Cellular Therapy Program, Division of Hematology/Oncology, Department of Internal Medicine, Greenebaum Comprehensive Cancer Center, University of Maryland Medical Center, University of Maryland School of Medicine, Baltimore, MD
| | - Hanan Alkhaldi
- Transplantation & Cellular Therapy Program, Division of Hematology/Oncology, Department of Internal Medicine, Greenebaum Comprehensive Cancer Center, University of Maryland Medical Center, University of Maryland School of Medicine, Baltimore, MD
| | - John Preston Claiborne
- Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, MD
| | - Seung Tae Lee
- Transplantation & Cellular Therapy Program, Division of Hematology/Oncology, Department of Internal Medicine, Greenebaum Comprehensive Cancer Center, University of Maryland Medical Center, University of Maryland School of Medicine, Baltimore, MD
| | - Kathryn Kline
- Transplantation & Cellular Therapy Program, Division of Hematology/Oncology, Department of Internal Medicine, Greenebaum Comprehensive Cancer Center, University of Maryland Medical Center, University of Maryland School of Medicine, Baltimore, MD
| | - Jennie Y Law
- Transplantation & Cellular Therapy Program, Division of Hematology/Oncology, Department of Internal Medicine, Greenebaum Comprehensive Cancer Center, University of Maryland Medical Center, University of Maryland School of Medicine, Baltimore, MD
| | - Jean A Yared
- Transplantation & Cellular Therapy Program, Division of Hematology/Oncology, Department of Internal Medicine, Greenebaum Comprehensive Cancer Center, University of Maryland Medical Center, University of Maryland School of Medicine, Baltimore, MD
| |
Collapse
|
6
|
Milunović V, Dragčević D, Bogeljić Patekar M, Mandac Smoljanović I, Gašparov S. The Improving Outcomes in Relapsed-Refractory Diffuse Large B Cell Lymphoma: The Role of CAR T-Cell Therapy. Curr Treat Options Oncol 2025; 26:445-464. [PMID: 40293655 DOI: 10.1007/s11864-025-01305-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 02/17/2025] [Indexed: 04/30/2025]
Abstract
OPINION STATEMENT Diffuse large B cell lymphoma, not otherwise specified (DLBCL-NOS) is the most common aggressive lymphoma and can be cured with CHOP-R immunochemotherapy in 60% of cases. The second-line therapy includes salvage regimens followed by autologous stem cell transplantation (ASCT), which offers a cure to a minority of patients due to limitations in efficacy and eligibility. These data present the unmet need in the field, and this review article focuses on how second-generation chimeric antigen receptor T (CAR T) cell therapy targeting CD19 antigen may improve the outcomes with relapsed/refractory DLBCL. In heavily pretreated patients, who have dismal outcomes with conventional therapy, all three approved products-tisangenlecleucel (tisa-cel), axicabtagene ciloleucel (axi-cel), and lisocabtagene maraleucel (liso-cel) have shown durable, unprecedented complete responses with the potential for cure. When compared to salvage regimens and ASCT as the standard of care, axi-cel and liso-cel, unlike tisa-cel, have demonstrated superiority in long-term control. In ASCT-ineligible r/r DLBCL, liso-cel has shown a favourable benefit-risk ratio. Regarding safety, two adverse events of interest have emerged: cytokine release syndrome and immune effector cell-associated neurotoxicity syndrome, both of which are manageable. Real-world evidence reflects the results of pivotal trials while favouring axi-cel in heavily pretreated patients, albeit with higher toxicity. The main barrier to the implementation of this treatment modality is the cost associated with the process of CAR T therapy, along with complications and reimbursement issues. However, the barriers can be overcome, and CAR T therapy has the potential to become the standard of care in relapsed/refractory DLBCL. Furthermore, with advances in the scientific engineering of CAR products and the understanding of novel treatment modalities currently being tested in clinical trials, we believe that targeted cellular therapy will become the future of relapsed/refractory DLBCL treatment.
Collapse
MESH Headings
- Humans
- Lymphoma, Large B-Cell, Diffuse/therapy
- Lymphoma, Large B-Cell, Diffuse/mortality
- Lymphoma, Large B-Cell, Diffuse/pathology
- Lymphoma, Large B-Cell, Diffuse/immunology
- Lymphoma, Large B-Cell, Diffuse/etiology
- Immunotherapy, Adoptive/methods
- Immunotherapy, Adoptive/adverse effects
- Receptors, Chimeric Antigen/immunology
- Treatment Outcome
- Salvage Therapy
- Combined Modality Therapy
- Antigens, CD19/immunology
- Drug Resistance, Neoplasm
- Disease Management
- Neoplasm Recurrence, Local/therapy
- Antineoplastic Combined Chemotherapy Protocols/therapeutic use
- Receptors, Antigen, T-Cell
- Clinical Trials as Topic
Collapse
Affiliation(s)
- Vibor Milunović
- Division of Hematology, Clinical Hospital Merkur, Zajčeva 19, 10000, Zagreb, Croatia.
| | - Dora Dragčević
- Division of Hematology, Clinical Hospital Merkur, Zajčeva 19, 10000, Zagreb, Croatia
| | | | | | - Slavko Gašparov
- School of Medicine in Zagreb, University of Zagreb, Zagreb, Croatia
- Clinical Department of Cytology and Pathology, Clinical Hospital Merkur, Zagreb, Croatia
| |
Collapse
|
7
|
Lakhotia R, Melani C, Dunleavy K, Pittaluga S, Desai S, Ahlman MA, Lucas N, Steinberg SM, Jaffe ES, Wilson WH, Roschewski M. Phase 2 study of alemtuzumab and dose-adjusted EPOCH-R in relapsed or refractory aggressive B-cell lymphomas. Leuk Lymphoma 2025; 66:1088-1099. [PMID: 39899393 DOI: 10.1080/10428194.2025.2457553] [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: 10/25/2024] [Revised: 01/12/2025] [Accepted: 01/18/2025] [Indexed: 02/05/2025]
Abstract
Immune cells within the lymphoma tumor microenvironment promote immune evasion and are rational therapeutic targets. Alemtuzumab targets CD52 expressed on malignant B-cells and infiltrating nonmalignant T-cells. We evaluated the safety and efficacy of alemtuzumab with DA-EPOCH-R in 48 patients with relapsed/refractory aggressive B-cell lymphoma. Febrile neutropenia occurred in 18% of cycles and serious infections in 21% of patients. Responses were observed in 30 (62%) patients, including 12 (80%) patients with classical HL and 3 (75%) patients with T-cell/histiocyte-rich large B-cell lymphoma (THRLCL). Seventeen (35%) patients achieved complete responses, and 12 (25%) were bridged to consolidation. The 2-year progression-free survival (PFS) and overall survival were 22.1% (95% CI, 11.5-34.7%) and 45.2% (95% CI, 34.3-58.9%), respectively. The 2-year PFS for HL and THRLCL patients was 35% and 50%, respectively. Alemtuzumab can be safely combined with DA-EPOCH-R in relapsed/refractory aggressive B-cell lymphomas and can induce durable responses in patients with T-cell-rich microenvironments.
Collapse
Affiliation(s)
- Rahul Lakhotia
- Lymphoid Malignancies Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - Christopher Melani
- Lymphoid Malignancies Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - Kieron Dunleavy
- Hematology, Georgetown Lombardi Comprehensive Cancer Center, Washington, DC, USA
| | - Stefania Pittaluga
- Laboratory of Pathology, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - Sanjal Desai
- Division of Hematology, Oncology, and Transplantation, University of Minnesota, Minneapolis, MN, USA
| | - Mark A Ahlman
- Radiology and Imaging, Medical College of Georgia, Augusta, GA, USA
| | - Nicole Lucas
- Pediatric Oncology Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - Seth M Steinberg
- Biostatistics and Data Management Section, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - Elaine S Jaffe
- Laboratory of Pathology, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - Wyndham H Wilson
- Lymphoid Malignancies Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - Mark Roschewski
- Lymphoid Malignancies Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| |
Collapse
|
8
|
Yu XJ, Liu C, Hu SZ, Yuan ZY, Ni HY, Sun SJ, Hu CY, Zhan HQ. Application of CAR-T cell therapy in B-cell lymphoma: a meta-analysis of randomized controlled trials. Clin Transl Oncol 2025; 27:2700-2709. [PMID: 39514165 DOI: 10.1007/s12094-024-03774-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2024] [Accepted: 10/18/2024] [Indexed: 11/16/2024]
Abstract
BACKGROUND This study aims to compare the efficacy and safety of chimeric antigen receptor T-cell (CAR-T) immunotherapy with standard treatment for B-cell lymphoma, providing evidence-based support for the more efficient use of CAR-T cell immunotherapy. METHODS We conducted a comprehensive literature search of high-quality randomized controlled trials (RCTs) on CAR-T therapy for B-cell lymphoma in the following databases: Wanfang, Web of Science, CNKI, VIP database, and PubMed, up to February 2024. The outcome measures included objective remission rate (ORR), complete remission rate (CRR), and incidence of adverse reactions. Subgroup analysis was performed based on the differences in co-stimulatory domains. Meta-analysis was conducted using Review Manager 5.4 and Stata software. RESULTS A total of five RCTs involving 1670 patients were included in this meta-analysis. The results showed that the CAR-T treatment group had significantly higher ORR (RR: 1.47, 95% CI 1.23-1.76, I2 = 80%, p < 0.0001), CRR (RR: 2.19, 95% CI 2.16-3.79, I2 = 93%, p = 0.005), cytokine release syndrome (CRS) incidence (RR: 34.51, 95% CI 2.27-523.78, I2 = 98%, p = 0.01), neurotoxicity (NT) incidence (RR: 6.00, 95% CI 1.82-19.75, I2 = 80%, p = 0.003), neutropenia incidence (RR: 1.39, 95% CI 1.02-1.88, I2 = 93%, p = 0.03), leukopenia incidence (RR: 1.39, 95% CI 1.04-1.87, I2 = 61%, p = 0.03), and headache incidence (RR: 1.56, 95% CI 1.25-1.95, I2 = 34%, p < 0.0001) compared to the standard treatment group. Subgroup analysis based on co-stimulatory domains revealed that the 4-1BB subgroup had higher incidences of CRR, CRS, NT and leukopenia than the CD28 subgroup; however, the CD28 subgroup exhibited higher ORR and neutropenia than the 4-1BB subgroup. CONCLUSION CAR-T cell immunotherapy demonstrates superior efficacy compared to standard therapy in treating B-cell lymphoma. However, CAR-T treatment can lead to adverse reactions such as CRS and NT. Infusion of an appropriate dose of CAR-T cells (e.g., 100 × 106) may be a strategy to mitigate the risk of CRS and NT.
Collapse
Affiliation(s)
- Xiao-Jing Yu
- Department of Pathology, The Second Affiliated Hospital of Anhui Medical University, Hefei, 230601, China
- Department of Pathology, School of Basic Medical Sciences, Anhui Medical University, Hefei, 230032, China
| | - Chang Liu
- Department of Pathology, School of Basic Medical Sciences, Anhui Medical University, Hefei, 230032, China
| | - Shi-Zhi Hu
- Department of Pathology, The Second Affiliated Hospital of Anhui Medical University, Hefei, 230601, China
| | - Zhan-Yuan Yuan
- Department of Plastic and Reconstructive Surgery, The Second Affiliated Hospital of Anhui Medical University, Hefei, 230601, China
| | - Hai-Yan Ni
- Department of Pathology, School of Basic Medical Sciences, Anhui Medical University, Hefei, 230032, China
| | - Sheng-Jia Sun
- Clinical Medical College of Anhui Medical University, Hefei, 230031, China
| | - Cheng-Yang Hu
- Department of Epidemiology and Biostatistics, Anhui Medical University, Hefei, 230032, China.
| | - He-Qin Zhan
- Department of Pathology, The Second Affiliated Hospital of Anhui Medical University, Hefei, 230601, China.
- Department of Pathology, School of Basic Medical Sciences, Anhui Medical University, Hefei, 230032, China.
| |
Collapse
|
9
|
Ohmachi K. JSH practical guidelines for hematological malignancies, 2023: II. Lymphoma5. Diffuse large B-cell lymphoma, not otherwise specified (DLBCL, NOS). Int J Hematol 2025:10.1007/s12185-025-03997-z. [PMID: 40434571 DOI: 10.1007/s12185-025-03997-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2025] [Revised: 04/10/2025] [Accepted: 04/24/2025] [Indexed: 05/29/2025]
Affiliation(s)
- Ken Ohmachi
- Department of Internal Medicine, Division of Hematology and Oncology, Tokai University School of Medicine, 143 Shimokasuya, Isehara-City, Kanagawa, 259-1143, Japan.
| |
Collapse
|
10
|
Juthani R, Pugh K, Neuendorff NR, Torka P. Representation of older adults in registrational trials associated with therapeutic approvals in diffuse large B-cell lymphoma. J Geriatr Oncol 2025; 16:102264. [PMID: 40412090 DOI: 10.1016/j.jgo.2025.102264] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2025] [Revised: 04/06/2025] [Accepted: 05/16/2025] [Indexed: 05/27/2025]
Affiliation(s)
- Ronit Juthani
- Department of Medicine, Saint Vincent Hospital, Worcester, MA, USA.
| | - Kelly Pugh
- Division of Hematology, James Cancer Center, The Ohio State University, Columbus, United States
| | - Nina Rosa Neuendorff
- Department of Geriatrics, Marien Hospital Herne, University Hospital Ruhr University Bochum, Herne, Germany
| | - Pallawi Torka
- Lymphoma Service, Memorial Sloan Kettering Cancer Center, New York City, United States
| |
Collapse
|
11
|
Robak E, Braun M, Robak T. A current view of the pathogenesis and treatment of primary cutaneous diffuse large B cell lymphoma - leg type. Leuk Lymphoma 2025:1-12. [PMID: 40393040 DOI: 10.1080/10428194.2025.2506503] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2025] [Revised: 05/05/2025] [Accepted: 05/10/2025] [Indexed: 05/22/2025]
Abstract
Primary cutaneous diffuse large B cell lymphoma, leg type (PCDLBCL-LT) is an aggressive B cell extranodal variant of lymphoma present in the skin, typically without evidence of extra cutaneous spread at the time of diagnosis. PCDLBCL-LT accounts for 20% of all primary cutaneous B cell lymphomas (CBCL) and 5% of all primary cutaneous lymphomas (PCL). It is more common in the elderly (median age 75 years). The disease commonly manifests as rapidly-growing red to bluish often ulcerating, nodular tumors, plaques or violaceous nodules on one or both lower extremities. Only 10% to 15% of lesions develop in other areas. A prognosis of PCDLBCL-LT is poor, with a 5-year survival rate of 40 to 50%. The first-line treatment of PCDLBCL-LT includes immunochemotherapy, most commonly R-CHOP (rituximab, cyclophosphamide, doxorubicin hydrochloride, vincristine sulfate, prednisone). In the case of a solitary lesion, excision or radiotherapy should be considered. Many patients demonstrate cutaneous relapses (70%) or systemic dissemination (50%). Recent studies have reported the use of Bruton's tyrosine-kinase (BTK) inhibitors, BCL2 inhibitors, immunomodulatory drugs and immune check-point inhibitors in treating relapsed or refractory patients. The study summarizes the current view of the pathogenesis, diagnosis and treatment of PCDLBCL-LT, including genetic abnormalities and novel targeted drugs.
Collapse
Affiliation(s)
- Ewa Robak
- Department of Dermatology, Medical University of Lodz, Lodz, Poland
| | - Marcin Braun
- Department of Pathology, Chair of Oncology, Medical University of Lodz, Lodz, Poland
| | - Tadeusz Robak
- Department of Hematology, Medical University of Lodz, Lodz, Poland
- Department of General Hematology and Internal Medicine, Copernicus Memorial Hospital, Lodz, Poland
| |
Collapse
|
12
|
Chaganti S, Fox CP, Maybury BD, Burton C, Barrington SF, Illidge T, Kalakonda N, Eyre TA, McKay P, Kuhnl A, Cwynarski K, Davies AJ. Management of relapsed or refractory large B-cell lymphoma: A British Society for Haematology Guideline. Br J Haematol 2025. [PMID: 40384597 DOI: 10.1111/bjh.20129] [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: 02/07/2025] [Accepted: 04/18/2025] [Indexed: 05/20/2025]
Abstract
Time to progression is the strongest predictor of outcome in relapsed diffuse large B-cell lymphoma. Second-line treatment with chimeric antigen receptor (CAR) T-cell therapy is recommended for patients with progression within 12 months of first-line chemoimmunotherapy. In patients with late relapse, platinum-based chemotherapy followed by high-dose chemotherapy with autologous stem cell rescue is recommended. In second relapse, CAR T-cell or CD3xCD20 bispecific antibody therapy is recommended in eligible patients. Other treatment options are available for less fit patients. Specific recommendations are made on diagnostic immunohistochemistry, bendamustine use and bridging to CAR T-cell therapy.
Collapse
Affiliation(s)
- Sridhar Chaganti
- Centre for Clinical Haematology, University Hospitals Birmingham, Birmingham, United Kingdom
| | - Christopher P Fox
- School of Medicine, University of Nottingham, Nottingham, United Kingdom
| | - Bernard D Maybury
- Centre for Clinical Haematology, University Hospitals Birmingham, Birmingham, United Kingdom
| | - Cathy Burton
- Department of Haematology, The Leeds Teaching Hospitals, Leeds, United Kingdom
| | - 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, London, United Kingdom
| | - Timothy Illidge
- Manchester NIHR Biomedical Research Centre, Division of Cancer Sciences, University of Manchester, Manchester, United Kingdom
| | - Nagesh Kalakonda
- Department of Molecular and Clinical Cancer Medicine, University of Liverpool, Liverpool, United Kingdom
| | - Toby A Eyre
- Oxford Cancer and Haematology Centre, Oxford University Hospitals, Oxford, United Kingdom
| | - Pam McKay
- Department of Haematology, Beatson West of Scotland Cancer Centre, Glasgow, United Kingdom
| | - Andrea Kuhnl
- Department of Haematology, King's College Hospital, London, United Kingdom
| | - Kate Cwynarski
- Department of Haematology, University College London Hospitals, London, United Kingdom
| | - Andrew J Davies
- Cancer Sciences Divisions, Centre for Cancer Immunology, University of Southampton, Southampton, United Kingdom
| |
Collapse
|
13
|
Hotchandani H, Sengar M, Shetty A, John A, Karulkar A, Kalra D, Ravikumar S, Jaiswal A, Yadav Y, Parkar S, Walawalkar A, Kadam S, Bagal B, Nayak L, Purwar R, Jain H. Implementation of a CAR-T cell therapy program in India. Expert Rev Anticancer Ther 2025:1-10. [PMID: 40336352 DOI: 10.1080/14737140.2025.2501744] [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: 01/09/2025] [Accepted: 04/30/2025] [Indexed: 05/09/2025]
Abstract
INTRODUCTION CAR-T cell therapy has changed the treatment paradigm for hematological malignancies, offering a curative potential for patients with relapsed or refractory disease. India, with its significant burden of hematological malignancies, faces unique challenges in implementing this therapy. The development of indigenous CAR-T cells has reduced costs substantially, but barriers remain, including limited manufacturing capacity, relatively high costs, and logistical constraints. AREAS COVERED This article emphasizes the importance of optimized patient selection and triaging of apheresis slots to maximize the benefits of CAR-T cell therapy. The use of efficient bridging therapies and antibody-based approaches are being explored to improve outcomes, particularly in aggressive lymphomas and leukemias. Opportunities lie in leveraging India's growing biotechnology sector for cost-efficient production and in evaluating novel combination therapies to enhance CAR-T cell efficacy. This article also explores the technical and socioeconomic challenges of CAR-T cell development in India and suggests strategies to enhance accessibility, affordability, and implementation. EXPERT OPINION Ongoing advancements and research may help tailor CAR-T cell protocols to the local population. Future integration of NK cell therapy, TCR-based approaches, and multi-antigen targeting holds promise for enhancing therapeutic efficacy.
Collapse
Affiliation(s)
- Hema Hotchandani
- Department of Medical Oncology, Tata Memorial Hospital, Mumbai, India
| | - Manju Sengar
- Department of Medical Oncology, Tata Memorial Hospital, Mumbai, India
| | - Alok Shetty
- Department of Medical Oncology, Tata Memorial Hospital, Mumbai, India
| | - Anupa John
- Department of Medical Oncology, Tata Memorial Hospital, Mumbai, India
| | - Atharva Karulkar
- ImmunoAdoptive Cell Therapy Private Limited (ImmunoACT Pvt Ltd), Mumbai, India
| | - Devanshi Kalra
- ImmunoAdoptive Cell Therapy Private Limited (ImmunoACT Pvt Ltd), Mumbai, India
| | - Smrithi Ravikumar
- ImmunoAdoptive Cell Therapy Private Limited (ImmunoACT Pvt Ltd), Mumbai, India
| | - Anjali Jaiswal
- ImmunoAdoptive Cell Therapy Private Limited (ImmunoACT Pvt Ltd), Mumbai, India
| | - Yuktam Yadav
- ImmunoAdoptive Cell Therapy Private Limited (ImmunoACT Pvt Ltd), Mumbai, India
| | | | | | - Sonali Kadam
- Nursing Department, Tata Memorial Hospital, Mumbai, India
| | - Bhausaheb Bagal
- Department of Medical Oncology, Tata Memorial Hospital, Mumbai, India
| | - Lingaraj Nayak
- Department of Medical Oncology, Tata Memorial Hospital, Mumbai, India
| | - Rahul Purwar
- ImmunoAdoptive Cell Therapy Private Limited (ImmunoACT Pvt Ltd), Mumbai, India
| | - Hasmukh Jain
- Department of Medical Oncology, Tata Memorial Hospital, Mumbai, India
| |
Collapse
|
14
|
Qi S, Li J, Gu X, Zhang Y, Zhou W, Wang F, Wang W. Impacts of ageing on the efficacy of CAR-T cell therapy. Ageing Res Rev 2025; 107:102715. [PMID: 40058461 DOI: 10.1016/j.arr.2025.102715] [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: 01/14/2025] [Revised: 02/19/2025] [Accepted: 03/02/2025] [Indexed: 03/17/2025]
Abstract
Chimeric antigen receptor T cells recognizing CD19 (19CAR-T) cell therapy has achieved robust clinical efficacy when treating some hematological malignancies, but which patient subgroups benefit mostly remains elusive. Here we summarized the data of 541 patients from 30 clinical trials who underwent 19 CAR-T therapy and analyzed the different clinical responses between young (<44 years), middle-aged (45-59 years) and elderly (>60 years) patients and found that the young patients showed a higher level of complete response (CR) rate. Therefore, we then summarize the advances of studies focusing on the effects of age on anti-tumor efficacy of CAR-T therapy and analyze the reasons for the low CR rate after CAR-T cell therapy in elderly patients with tumors, aiming to provide hints for oncologists to select the most suitable candidate for this cancer immunotherapy.
Collapse
Affiliation(s)
- Shimao Qi
- Department of Biotherapy, State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Collaborative Innovation Center for Biotherapy, Sichuan University, Chengdu 610041, PR China
| | - Jiaqian Li
- Department of Biotherapy, State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Collaborative Innovation Center for Biotherapy, Sichuan University, Chengdu 610041, PR China
| | - Xinyu Gu
- Department of Biotherapy, State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Collaborative Innovation Center for Biotherapy, Sichuan University, Chengdu 610041, PR China
| | - Yalan Zhang
- Department of Biotherapy, State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Collaborative Innovation Center for Biotherapy, Sichuan University, Chengdu 610041, PR China
| | - Weilin Zhou
- Department of Biotherapy, State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Collaborative Innovation Center for Biotherapy, Sichuan University, Chengdu 610041, PR China
| | - Fengling Wang
- Department of Biotherapy, State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Collaborative Innovation Center for Biotherapy, Sichuan University, Chengdu 610041, PR China
| | - Wei Wang
- Department of Biotherapy, State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Collaborative Innovation Center for Biotherapy, Sichuan University, Chengdu 610041, PR China.
| |
Collapse
|
15
|
Jamil A, Qureshi Z, Siddique R, Altaf F, Akram H, Jamil R, Aslam S, Selene II. Efficacy and Safety of Chimeric Antigen Receptor (CAR)-T Cell Therapy in Patients with Non-Hodgkin Lymphoma: A Systematic Review and Meta-Analysis. Am J Clin Oncol 2025; 48:262-270. [PMID: 39924687 DOI: 10.1097/coc.0000000000001171] [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] [Indexed: 02/11/2025]
Abstract
OBJECTIVES Non-Hodgkin lymphomas (NHL) are a diverse group of lymphoproliferative malignancies, often more unpredictable than Hodgkin lymphomas, with a higher likelihood of extranodal spread. NHL's resistance to standard chemotherapy has increased, leading to a growing interest in personalized treatments like chimeric antigen receptor T-cell therapies (CAR-TCT). METHODS A literature search was conducted across PubMed, ScienceDirect, Google Scholar, and the Cochrane Library for studies on CAR-TCT in NHL treatment published until July 2024. The outcomes assessed included overall survival (OS), event-free survival (EFS), progression-free survival (PFS), objective response rate (ORR), and adverse events (AEs). Data were pooled using RevMan 5.41 and Comprehensive Meta-analysis 3. RESULTS Out of 532 articles, 8 met the inclusion criteria. CAR-TCT significantly improved OS (HR: 0.79; 95% CI: 0.63-1.00; P =0.05) and PFS (HR: 0.46; 95% CI: 0.36-0.58; P <0.00001) compared with standard chemotherapy. However, EFS was not significantly different (HR: 0.54; 95% CI: 0.26-1.09; P =0.09). About 76.6% of NHL patients responded to CAR-TCT, but the ORR was similar between CAR-TCT and standard therapy (MD: 19.23%; 95% CI: -11.34% to 49.80%; P =0.22). Safety analysis found a grade ≥3 AEs incidence comparable to CAR-TCT and standard care. However, CAR-TCT was associated with higher neutropenia risk but lower thrombocytopenia, anemia, and nausea risks. CONCLUSION CAR-TCT significantly improves OS and PFS in refractory NHL but does not notably impact EFS. While its ORR is comparable to standard chemotherapy, CAR-TCT has a better safety profile, making it a promising treatment option.
Collapse
Affiliation(s)
- Abdur Jamil
- Department of Medicine, Samaritan Medical Centre Watertown
| | - Zaheer Qureshi
- Assistant Professor of Medicine, The Frank H. Netter M.D. School of Medicine at Quinnipiac University, Bridgeport, CT
| | | | - Faryal Altaf
- Department of Internal Medicine, Icahn School of Medicine at Mount Sinai/BronxCare Health System, New York, NY
| | | | - Rohma Jamil
- FMH College of Medicine and Dentistry, Lahore, Pakistan
| | - Shehroz Aslam
- Department of Internal Medicine, Core Faculty, Samaritan Medical Center, Watertown
| | | |
Collapse
|
16
|
Yamamoto C, Honda S, Tominaga R, Yokoyama D, Furuki S, Noguchi A, Koyama S, Murahashi R, Nakashima H, Kawaguchi SI, Hyodo K, Toda Y, Umino K, Minakata D, Ashizawa M, Ueda M, Hatano K, Sato K, Ohmine K, Fujiwara SI, Kanda Y. Impact of Real-World Clinical Factors on an Analysis of the Cost-Effectiveness of "Immediate CAR-T" Versus "Late CAR-T" as Second-Line Treatment for DLBCL Patients. Transplant Cell Ther 2025; 31:339.e1-339.e15. [PMID: 39954961 DOI: 10.1016/j.jtct.2025.02.013] [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: 10/18/2024] [Revised: 02/06/2025] [Accepted: 02/10/2025] [Indexed: 02/17/2025]
Abstract
While chimeric antigen receptor (CAR-T) targeting CD19 as second-line therapy for diffuse large B cell lymphoma (DLBCL) is a promising strategy, the high costs and limited access to CAR-T pose significant challenges. When assessing the cost-effectiveness of CAR-T, we need to consider not only individual outcomes but also how to effectively integrate CAR-T into the overall treatment approach for relapsed DLBCL. We conducted a cost-effective analysis for patients with DLBCL in early relapse or primary refractory, to compare "immediate CAR-T," which proceeds directly to CAR-T, and "late CAR-T," which initially aims at ASCT and quickly switches to third-line CAR-T if non-responsive. The primary analysis used a patient age of 60 years, and it also examined variations from 40 to 70 years. The analysis was performed for both Japanese and US settings using a Markov model incorporating life expectancy in both countries, with extensive sensitivity analysis including factors such as age, the choice of CAR-T (lisocabtagene maraleucel or axicabtagene ciloleucel), and the opportunity to receive third-line CAR-T, to reflect real-world situations. The length of a Markov cycle was defined to be 1 month, and patients in the model were assumed to age 1 year every 12 Markov cycles. The analysis was made over a lifetime horizon, and the outcome was measured based on incremental cost-effectiveness ratio (ICER), with willingness-to-pay (WTP) thresholds of ¥7,500,000 and $150,000 per quality-adjusted life years (QALY) in Japan and the US, respectively, with an annual discount rate of 3%. Compared with "late CAR-T," the "immediate CAR-T" strategy gained QALYs of 0.97 and 0.89 with an incremental cost of ¥5,998,354 and $88,440 in Japan and the US, respectively. The ICERs were ¥6,170,058/QALY in Japan and $99,596/QALY in the US. In the probabilistic sensitivity analysis for patients aged 60, "immediate CAR-T" was cost-effective in 54.8% and 61.7% of the 10,000 Monte Carlo iterations in Japan and the US, respectively. Sensitivity analyses showed that "immediate CAR-T" was not cost-effective when patients were over 68.4 in Japan, when the standardized mortality ratio of CAR-T and ASCT survivors was close, and when utility during treatment-free remission was low. Incorporating various clinical factors, the analysis showed that "immediate CAR-T" is more cost-effective than "late CAR-T." However, this conclusion should be interpreted with caution, as the ICERs were very close to the WTP thresholds, and the results were highly sensitive to parameter changes.
Collapse
Affiliation(s)
- Chihiro Yamamoto
- Division of Hematology, Department of Medicine, Jichi Medical University, Shimotsuke, Japan
| | - Seina Honda
- Division of Hematology, Department of Medicine, Jichi Medical University, Shimotsuke, Japan
| | - Ryutaro Tominaga
- Division of Hematology, Department of Medicine, Jichi Medical University, Shimotsuke, Japan
| | - Daizo Yokoyama
- Division of Hematology, Department of Medicine, Jichi Medical University, Shimotsuke, Japan
| | - Shuka Furuki
- Division of Hematology, Department of Medicine, Jichi Medical University, Shimotsuke, Japan
| | - Atsuto Noguchi
- Division of Hematology, Department of Medicine, Jichi Medical University, Shimotsuke, Japan
| | - Shunsuke Koyama
- Division of Hematology, Department of Medicine, Jichi Medical University, Shimotsuke, Japan
| | - Rui Murahashi
- Division of Hematology, Department of Medicine, Jichi Medical University, Shimotsuke, Japan
| | - Hirotomo Nakashima
- Division of Hematology, Department of Medicine, Jichi Medical University, Shimotsuke, Japan
| | - Shin-Ichiro Kawaguchi
- Division of Hematology, Department of Medicine, Jichi Medical University, Shimotsuke, Japan
| | - Kazuki Hyodo
- Division of Hematology, Department of Medicine, Jichi Medical University, Shimotsuke, Japan
| | - Yumiko Toda
- Division of Hematology, Department of Medicine, Jichi Medical University, Shimotsuke, Japan
| | - Kento Umino
- Division of Hematology, Department of Medicine, Jichi Medical University, Shimotsuke, Japan
| | - Daisuke Minakata
- Division of Hematology, Department of Medicine, Jichi Medical University, Shimotsuke, Japan
| | - Masahiro Ashizawa
- Division of Hematology, Department of Medicine, Jichi Medical University, Shimotsuke, Japan
| | - Masuzu Ueda
- Division of Hematology, Department of Medicine, Jichi Medical University, Shimotsuke, Japan
| | - Kaoru Hatano
- Division of Hematology, Department of Medicine, Jichi Medical University, Shimotsuke, Japan
| | - Kazuya Sato
- Division of Hematology, Department of Medicine, Jichi Medical University, Shimotsuke, Japan
| | - Ken Ohmine
- Division of Hematology, Department of Medicine, Jichi Medical University, Shimotsuke, Japan
| | - Shin-Ichiro Fujiwara
- Division of Hematology, Department of Medicine, Jichi Medical University, Shimotsuke, Japan; Division of Cell Transplantation and Transfusion, Jichi Medical University, Shimotsuke, Japan
| | - Yoshinobu Kanda
- Division of Hematology, Department of Medicine, Jichi Medical University, Shimotsuke, Japan.
| |
Collapse
|
17
|
Gazeau N, Beauvais D, Tilmont R, Srour M, Ferrant E, Safar V, Fouillet L, Flandrin-Gresta P, Gower N, Chauvet P, Duployez N, Podvin B, Demaret J, Huet S, Sujobert P, Ghesquières H, Damaj G, Bachy E, Morschhauser F, Yakoub-Agha I, Heiblig M, Sesques P. Myeloid neoplasms after CD19-directed CAR T cells therapy in long-term B-cell lymphoma responders, a rising risk over time? Leukemia 2025:10.1038/s41375-025-02605-7. [PMID: 40275069 DOI: 10.1038/s41375-025-02605-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2025] [Revised: 03/09/2025] [Accepted: 04/02/2025] [Indexed: 04/26/2025]
Abstract
Therapy-related myeloid neoplasms (t-MN), including myelodysplastic neoplasms (t-MDS) and acute myeloid leukemia (t-AML), have emerged as significant late complications after CAR T cell therapy. We retrospectively analyzed 539 patients with B cell lymphoma treated with CD19 directed CAR T cell therapy across four French centers. Cumulative incidences of t-MN was estimated with relapse or death treated as competing risk. Univariate and propensity score matching (PSM) analyses were conducted to assess risk factors with age and the number of prior treatments as covariates. After a median follow-up of 25 months, the cumulative incidence of t-MN was 4.5% at 2 years. T-MN occurred predominantly as t-MDS (62%) and t-AML (38%) with high cytogenetic risk. Median overall survival after t-MN diagnosis was 4.5 months. In univariate analysis, older age (p < 0.01), higher MCV (p < 0.01), and higher ICANS grade (p = 0.04) were associated with increased risk of t-MN. After PSM, MCV and ICANS grade remained significant risk factors. CAR T cell products with CD28 co-stimulatory domains trended towards higher t-MN risk (p = 0.09). NGS analysis showed that 85.7% of t-MN had pre-existing mutations, most commonly TP53. This study highlights t-MN as a severe late complication of CAR T cell therapy. MCV and ICANS grade were identified as key risk factors.
Collapse
Affiliation(s)
- Nicolas Gazeau
- Hematology Department, Centre Hospitalier Universitaire de Lille, Lille, France.
| | - David Beauvais
- Hematology Department, Centre Hospitalier Universitaire de Lille, Lille, France
| | - Rémi Tilmont
- Hematology Department, Centre Hospitalier Universitaire de Lille, Lille, France
| | - Micha Srour
- Hematology Department, Centre Hospitalier Universitaire de Lille, Lille, France
| | - Emmanuelle Ferrant
- Hematology Department, Hospices Civils de Lyon, Pierre Bénite, Lyon, France
| | - Violaine Safar
- Hematology Department, Hospices Civils de Lyon, Pierre Bénite, Lyon, France
| | - Ludovic Fouillet
- Hematology Department, Centre Hospitalier Universitaire de Saint-Etienne, Saint-Etienne, France
| | | | - Nicolas Gower
- Hematology Department, Centre Hospitalier Universitaire de Lille, Lille, France
| | - Paul Chauvet
- Hematology Department, Centre Hospitalier Universitaire de Lille, Lille, France
- CHU de Lille, Université de Lille, Inserm UMR1277, CNRS UMR9020-CANTHER, Lille, France
| | - Nicolas Duployez
- Biology and Pathology Center, Laboratory of Hematology, Centre Hospitalier Universitaire de Lille, Lille, France
| | - Benjamin Podvin
- Biology and Pathology Center, Laboratory of Hematology, Centre Hospitalier Universitaire de Lille, Lille, France
| | - Julie Demaret
- Biology and Pathology Center, Laboratory of Immunology, Centre Hospitalier Universitaire de Lille, Lille, France
| | - Sarah Huet
- Laboratory of Hematology, Centre Hospitalier Universitaire Lyon-Sud, Hospices Civils de Lyon, Pierre-Benite, France
| | - Pierre Sujobert
- Laboratory of Hematology, Centre Hospitalier Universitaire Lyon-Sud, Hospices Civils de Lyon, Pierre-Benite, France
| | - Hervé Ghesquières
- Hematology Department, Hospices Civils de Lyon, Pierre Bénite, Lyon, France
| | - Gandhi Damaj
- Hematology Department, Centre Hospitalier Universitaire de Caen, Caen, France
| | - Emmanuel Bachy
- Hematology Department, Hospices Civils de Lyon, Pierre Bénite, Lyon, France
| | - Franck Morschhauser
- Hematology Department, Centre Hospitalier Universitaire de Lille, Lille, France
| | - Ibrahim Yakoub-Agha
- Hematology Department, Centre Hospitalier Universitaire de Lille, Lille, France
- CHU de Lille, Université de Lille, INSERM U1286, Infinite, 59000, Lille, France
| | - Maël Heiblig
- Hematology Department, Hospices Civils de Lyon, Pierre Bénite, Lyon, France
| | - Pierre Sesques
- Hematology Department, Hospices Civils de Lyon, Pierre Bénite, Lyon, France
| |
Collapse
|
18
|
Galeano S, Bonfim C, Karduss A, Jaimovich G, Gómez-De León A, Bettarello G, Simione A, Correa C, Baldomero H, Neumann D, Basquiera AL, Berro M, Remaggi G, Amaru A, Barroso F, Seber A, Barriga F, Palma J, Puga B, Sánchez M, Herrera JM, Hernández C, Gómez-Almaguer D, Gaytán Morales F, Ruiz-Argüelles GJ, Mendoza N, Benítez ML, Wong A, Pagés C, Hernández M, Niederwieser D, Rondelli D, Frutos C. Results of the Latin American Bone Marrow Transplantation Society (LABMT) activity survey 2019-2022: the impact of the COVID-19 pandemic and the increase in related haploidentical donors. Bone Marrow Transplant 2025:10.1038/s41409-025-02600-7. [PMID: 40251416 DOI: 10.1038/s41409-025-02600-7] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2024] [Revised: 03/23/2025] [Accepted: 04/08/2025] [Indexed: 04/20/2025]
Abstract
A total of 6767 first hematopoietic cell transplants (HCT), 4121 autologous (61%) and 2646 allogeneic (39%), were reported by 166 teams from 12 Latin American countries that answered the 2022 LABMT/WBMT activity survey. The transplant rate (TR) for Latin America in 2022 was 103 HCT/10 million inhabitants with a wide variation between the different countries. The main indication for allogeneic (allo)-HCT was acute lymphoblastic leukaemia (41%) for the pediatric population and acute myeloid leukemia (32%) for adults. The main indication for autologous (auto)-HCT was neuroblastoma (33%) in children and plasma cell disorders (57%) in adults. In alloHCT, the most used hematopoietic cell source was the bone marrow (54%) in pediatric while peripheral blood stem cells (PBSC) (87%) was in adults. PBSC was the source of choice for autoHCT in both ages. The main trends observed in the period 2019-2022 was a decrease in the number of procedures in 2020 in association with the start of the COVID-19 pandemic, resuming growth in the following years. AlloHCT had a greater growth compared to autoHCT, and it was mainly driven by the utilization of haploidentical related donors, which became the main source from 2020 onwards.
Collapse
Affiliation(s)
| | | | | | | | - Andrés Gómez-De León
- Universidad Autónoma de Nuevo León, Facultad de Medicina y Hospital Universitario Dr. José Eleuterio González, Monterrey, México
| | | | | | | | - Helen Baldomero
- The WBMT Transplant Activity Survey Office, University Hospital, Basel, Switzerland
| | - Daniel Neumann
- Institute of Medical Informatics, Statistics and Epidemiology, University of Leipzig, Leipzig, Germany
| | - Ana Lisa Basquiera
- Hospital Privado Universitario de Córdoba, Instituto Universitario de Ciencias Biomédicas de Córdoba, Córdoba, Argentina
| | | | | | | | | | - Adriana Seber
- Hospital Samaritano Higienópolis and Instituto de Oncologia Pediátrica -Graacc/Universidade Federal de Sao Paulo, São Paulo, Brazil
| | | | - Julia Palma
- Comité Nacional de Trasplante de Médula Ósea-PINDA, Santiago, Chile
| | | | | | | | | | - David Gómez-Almaguer
- Universidad Autónoma de Nuevo León, Facultad de Medicina y Hospital Universitario Dr. José Eleuterio González, Monterrey, México
| | | | | | | | | | - Alfredo Wong
- Hospital Nacional Edgardo Rebagliati Martins, Lima, Perú
| | | | | | | | | | | |
Collapse
|
19
|
Furqan F, Ahn KW, Kaur M, Patel J, Ansell S, Awan FT, Baird J, Bezerra E, Farooq U, Fung H, Khurana A, Lekakis L, Lutfi F, McCarty J, Mukherjee A, Nath R, Romancik J, Schuster SJ, Smith M, Winter A, Turtle C, Sauter C, Shadman M, Herrara A, Hamadani M. Autologous Transplant or CAR-T as Consolidation Options in MYC Rearranged Large B-Cell Lymphoma Patients in Remission After Salvage Treatments. Am J Hematol 2025. [PMID: 40231369 DOI: 10.1002/ajh.27687] [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: 01/27/2025] [Revised: 03/31/2025] [Accepted: 04/02/2025] [Indexed: 04/16/2025]
Abstract
Although recent studies have demonstrated the efficacy of chimeric antigen receptor T-cell (CAR-T) therapy in relapsed large B-cell lymphoma (LBCL) with MYC rearrangement (R-MYC), the data comparing CAR-T to autologous hematopoietic cell transplant (auto-HCT) in such patients who achieve a complete or partial response (CR/PR) after salvage therapies are limited. We compared the clinical outcomes of patients with R-MYC LBCL (including double and triple hit lymphomas) who underwent CAR-T or auto-HCT after achieving a CR/PR with salvage therapies using the Center for International Blood & Marrow Transplant Research registry. Among the 252 patients (auto-HCT = 98, CAR-T = 154), relative to auto-HCT, CAR-T was associated with significantly lower overall survival (OS) (Hazard Ratio [HR] 2.09, 95% CI 1.38-3.15, p < 0.001) on multivariate analysis. There were no differences in progression-free survival (PFS) (HR 1.21, 95% CI 0.81-1.8 p = 0.36), risk of relapse (HR 1.1, 95% CI 0.71-1.69 p = 0.68), nonrelapse mortality (NRM) (HR 1.74, 95% CI 0.64-4.7 p = 0.28) while the post-relapse survival was longer in auto-HCT relative to CAR-T (HR 1.93, 95% CI 1.21-3.06 p = 0.01). On propensity score matched analysis accounting for differences in characteristics across the two cohorts, we detected no significant differences in OS (HR 1.72, 95% CI 0.92-3.21 p = 0.09), PFS (HR 1.04, 95% CI 0.64-1.68 p = 0.88), NRM (HR 1.22, 95% CI 0.35-4.2 p = 0.76), relapse (HR = 0.93, 95% CI 0.54-1.6 p = 0.8) and post-relapse survival (HR 2.25, 95% CI 0.98-5.17, p = 0.06). These data, although retrospective, support consideration for auto-HCT in patients with R-MYC LBCL who achieve a CR/PR after salvage therapies, particularly in regions with no or limited access to CAR-T.
Collapse
Affiliation(s)
- Fateeha Furqan
- BMT & Cellular Therapy Program, Department of Medicine, Medical College of Wisconsin, Milwaukee, Wisconsin, USA
| | - Kwang W Ahn
- Center for International Blood and Marrow Transplant Research, Department of Medicine, Medical College of Wisconsin, Milwaukee, Wisconsin, USA
- Division of Biostatistics, Institute for Health and Equity, Medical College of Wisconsin, Milwaukee, Wisconsin, USA
| | - Manmeet Kaur
- Center for International Blood and Marrow Transplant Research, Department of Medicine, Medical College of Wisconsin, Milwaukee, Wisconsin, USA
| | - Jinalben Patel
- Center for International Blood and Marrow Transplant Research, Department of Medicine, Medical College of Wisconsin, Milwaukee, Wisconsin, USA
| | - Stephen Ansell
- Hematology Division, Department of Internal Medicine, Mayo Clinic, Rochester, Minnesota, USA
| | - Farrukh T Awan
- Harold C. Simmons Comprehensive Cancer Center, University of Texas Southwestern Medical Center, Dallas, Texas, USA
| | - John Baird
- Division of Lymphoma, Department of Hematology and Hematopoietic Cell Transplantation, City of Hope, Duarte, California, USA
| | - Evandro Bezerra
- Division of Hematology, Department of Internal Medicine, James Comprehensive Cancer Center, The Ohio State University, Columbus, Ohio, USA
| | - Umar Farooq
- Department of Medicine, University of Iowa, Iowa City, Iowa, USA
| | - Henry Fung
- Department of Bone Marrow Transplant and Cellular Therapy at Fox Chase Cancer Center, Temple University, Philadelphia, Pennsylvania, USA
| | - Arushi Khurana
- Hematology Division, Department of Internal Medicine, Mayo Clinic, Rochester, Minnesota, USA
| | - Lazaros Lekakis
- Division of Transplantation and Cellular Therapy, University of Miami Hospital and Clinics, Sylvester Comprehensive Cancer Center, Miami, Florida, USA
| | - Forat Lutfi
- Division of Hematologic Malignancies & Cellular Therapeutics, University of Kansas Cancer Center, Westwood, Kansas, USA
| | - John McCarty
- Massey Cancer Center Bone Marrow Transplant Program, Virginia Commonwealth University, Richmond, Virginia, USA
| | | | - Rajneesh Nath
- Banner MD Anderson Cancer Center, Gilbert, Arizona, USA
| | - Jason Romancik
- Department of Hematology and Medical Oncology, Winship Cancer Institute at Emory University, Atlanta, Georgia, USA
| | - Stephen J Schuster
- Lymphoma Program, Abramson Cancer Center at University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Melody Smith
- Division of Blood and Marrow Transplantation and Cellular Therapy, Department of Medicine, Stanford University School of Medicine, Stanford, California, USA
| | - Allison Winter
- Department of Hematology and Medical Oncology, Taussig Cancer Institute, Cleveland Clinic, Cleveland, Ohio, USA
| | - Cameron Turtle
- Division of Hematology and Medical Oncology, University of Washington, Seattle, WA, USA
| | - Craig Sauter
- Department of Hematology and Medical Oncology, Taussig Cancer Institute, Cleveland Clinic, Cleveland, Ohio, USA
| | - Mazyar Shadman
- Division of Hematology and Medical Oncology, University of Washington, Seattle, WA, USA
| | - Alex Herrara
- Division of Lymphoma, Department of Hematology and Hematopoietic Cell Transplantation, City of Hope, Duarte, California, USA
| | - Mehdi Hamadani
- BMT & Cellular Therapy Program, Department of Medicine, Medical College of Wisconsin, Milwaukee, Wisconsin, USA
- Center for International Blood and Marrow Transplant Research, Department of Medicine, Medical College of Wisconsin, Milwaukee, Wisconsin, USA
| |
Collapse
|
20
|
Jadlowsky JK, Hexner EO, Marshall A, Grupp SA, Frey NV, Riley JL, Veloso E, McConville H, Rogal W, Czuczman C, Hwang WT, Li Y, Leskowitz RM, Farrelly O, Karar J, Christensen S, Barber-Rotenberg J, Gaymon A, Aronson N, Bernstein W, Melenhorst JJ, Roche AM, Everett JK, Zolnoski SA, McFarland AG, Reddy S, Petrichenko A, Cook EJ, Lee C, Gonzalez VE, Alexander K, Kulikovskaya I, Ramírez-Fernández Á, Minehart JC, Ruella M, Gill SI, Schuster SJ, Cohen AD, Garfall AL, Shah PD, Porter DL, Maude SL, Levine BL, Siegel DL, Chew A, McKenna S, Lledo L, Davis MM, Plesa G, Herbst F, Stadtmauer EA, Tebas P, DiNofia A, Haas A, Haas NB, Myers R, O'Rourke DM, Svoboda J, Tanyi JL, Aplenc R, Jacobson JM, Ko AH, Cohen RB, June CH, Bushman FD, Fraietta JA. Long-term safety of lentiviral or gammaretroviral gene-modified T cell therapies. Nat Med 2025; 31:1134-1144. [PMID: 39833408 DOI: 10.1038/s41591-024-03478-6] [Citation(s) in RCA: 10] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2024] [Accepted: 12/19/2024] [Indexed: 01/22/2025]
Abstract
Long-term risks of gene therapy are not fully understood. In this study, we evaluated safety outcomes in 783 patients over more than 2,200 total patient-years of observation from 38 T cell therapy trials. The trials employed integrating gammaretroviral or lentiviral vectors to deliver engineered receptors to target HIV-1 infection or cancer. Eighteen patients (2.3%) developed secondary malignancies after treatment, with a median onset of 1.94 years (range: 51 d to 14 years). Where possible, incident tumor samples were analyzed for vector copy number, revealing no evidence of high-level marking or other indications of insertional mutagenesis. One T cell lymphoma was detected, but malignant T cells were not marked by vector integration. Analysis of vector integration sites in 176 patients revealed no pathological insertions linked to secondary malignancies, although, in some cases, integration in or near specific genes, including tumor suppressor genes, was associated with modest clonal expansion and sustained T cell persistence. These findings highlight the safety of engineered T cell therapies.
Collapse
Affiliation(s)
- Julie K Jadlowsky
- Center for Cellular Immunotherapies, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Elizabeth O Hexner
- Center for Cellular Immunotherapies, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
- Department of Medicine, Division of Hematology/Oncology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Amy Marshall
- Center for Cellular Immunotherapies, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Stephan A Grupp
- Department of Pediatrics, Division of Oncology, Children's Hospital of Philadelphia, Philadelphia, PA, USA
| | - Noelle V Frey
- Department of Medicine, Division of Hematology/Oncology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
- Abramson Cancer Center, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - James L Riley
- Center for Cellular Immunotherapies, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
- Department of Microbiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Elizabeth Veloso
- Center for Cellular Immunotherapies, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Holly McConville
- Center for Cellular Immunotherapies, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Walter Rogal
- Center for Cellular Immunotherapies, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Cory Czuczman
- Center for Cellular Immunotherapies, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Wei-Ting Hwang
- Abramson Cancer Center, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
- Department of Biostatistics, Epidemiology, and Informatics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Yimei Li
- Department of Pediatrics, Division of Oncology, Children's Hospital of Philadelphia, Philadelphia, PA, USA
- Abramson Cancer Center, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
- Department of Biostatistics, Epidemiology, and Informatics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Rachel M Leskowitz
- Center for Cellular Immunotherapies, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Olivia Farrelly
- Center for Cellular Immunotherapies, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Jayashree Karar
- Center for Cellular Immunotherapies, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Shannon Christensen
- Center for Cellular Immunotherapies, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Julie Barber-Rotenberg
- Center for Cellular Immunotherapies, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Avery Gaymon
- Center for Cellular Immunotherapies, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Naomi Aronson
- Department of Medicine, Division of Infectious Diseases, Uniformed Services University of the Health Sciences, Bethesda, MD, USA
| | - Wendy Bernstein
- Department of Medicine, Division of Infectious Diseases, Uniformed Services University of the Health Sciences, Bethesda, MD, USA
| | - Jan Joseph Melenhorst
- Center for Cellular Immunotherapies, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
- Lerner Research Institute, Cleveland Clinic, Cleveland, OH, USA
| | - Aoife M Roche
- Department of Microbiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - John K Everett
- Department of Microbiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Sonja A Zolnoski
- Department of Microbiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Alexander G McFarland
- Department of Microbiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Shantan Reddy
- Department of Microbiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Angelina Petrichenko
- Department of Microbiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Emma J Cook
- Department of Microbiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Carole Lee
- Department of Microbiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Vanessa E Gonzalez
- Center for Cellular Immunotherapies, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Kathleen Alexander
- Center for Cellular Immunotherapies, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Irina Kulikovskaya
- Center for Cellular Immunotherapies, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Ángel Ramírez-Fernández
- Center for Cellular Immunotherapies, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
- Abramson Cancer Center, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
- Department of Microbiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Janna C Minehart
- Department of Medicine, Division of Hematology/Oncology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
- Abramson Cancer Center, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Marco Ruella
- Center for Cellular Immunotherapies, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
- Department of Medicine, Division of Hematology/Oncology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
- Abramson Cancer Center, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
- Parker Institute for Cancer Immunotherapy, University of Pennsylvania, Philadelphia, PA, USA
| | - Saar I Gill
- Center for Cellular Immunotherapies, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
- Department of Medicine, Division of Hematology/Oncology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
- Abramson Cancer Center, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
- Parker Institute for Cancer Immunotherapy, University of Pennsylvania, Philadelphia, PA, USA
| | - Stephen J Schuster
- Department of Medicine, Division of Hematology/Oncology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
- Abramson Cancer Center, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Adam D Cohen
- Department of Medicine, Division of Hematology/Oncology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
- Abramson Cancer Center, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Alfred L Garfall
- Department of Medicine, Division of Hematology/Oncology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
- Abramson Cancer Center, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Payal D Shah
- Department of Medicine, Division of Hematology/Oncology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
- Abramson Cancer Center, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
- Basser Center for BRCA, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - David L Porter
- Department of Medicine, Division of Hematology/Oncology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
- Abramson Cancer Center, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Shannon L Maude
- Center for Cellular Immunotherapies, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
- Department of Pediatrics, Division of Oncology, Children's Hospital of Philadelphia, Philadelphia, PA, USA
| | - Bruce L Levine
- Center for Cellular Immunotherapies, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
- Department of Pathology and Laboratory Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Donald L Siegel
- Center for Cellular Immunotherapies, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
- Abramson Cancer Center, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
- Department of Pathology and Laboratory Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Anne Chew
- Center for Cellular Immunotherapies, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
- Department of Pathology and Laboratory Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Stephen McKenna
- Center for Cellular Immunotherapies, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Lester Lledo
- Center for Cellular Immunotherapies, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Megan M Davis
- Center for Cellular Immunotherapies, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
- Department of Pathology and Laboratory Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Gabriela Plesa
- Center for Cellular Immunotherapies, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Friederike Herbst
- Center for Cellular Immunotherapies, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
- Department of Pathology and Laboratory Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Edward A Stadtmauer
- Department of Medicine, Division of Hematology/Oncology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
- Abramson Cancer Center, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Pablo Tebas
- Department of Medicine, Division of Infectious Diseases, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Amanda DiNofia
- Department of Pediatrics, Division of Oncology, Children's Hospital of Philadelphia, Philadelphia, PA, USA
| | - Andrew Haas
- Department of Medicine, Division of Pulmonary, Allergy and Critical Care, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Naomi B Haas
- Department of Medicine, Division of Hematology/Oncology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
- Abramson Cancer Center, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Regina Myers
- Department of Pediatrics, Division of Oncology, Children's Hospital of Philadelphia, Philadelphia, PA, USA
| | - Donald M O'Rourke
- Center for Cellular Immunotherapies, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
- Abramson Cancer Center, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
- Department of Neurosurgery, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Jakub Svoboda
- Department of Medicine, Division of Hematology/Oncology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
- Abramson Cancer Center, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Janos L Tanyi
- Abramson Cancer Center, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
- Department of Obstetrics and Gynecology, Division of Gynecologic Oncology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Richard Aplenc
- Department of Pediatrics, Division of Oncology, Children's Hospital of Philadelphia, Philadelphia, PA, USA
| | - Jeffrey M Jacobson
- Department of Medicine, Case Western Reserve University, Cleveland, OH, USA
| | - Andrew H Ko
- Department of Medicine, Division of Hematology/Oncology, University of California, San Francisco, San Francisco, CA, USA
| | - Roger B Cohen
- Department of Medicine, Division of Hematology/Oncology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
- Abramson Cancer Center, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Carl H June
- Center for Cellular Immunotherapies, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA.
- Abramson Cancer Center, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA.
- Parker Institute for Cancer Immunotherapy, University of Pennsylvania, Philadelphia, PA, USA.
- Department of Pathology and Laboratory Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA.
| | - Frederic D Bushman
- Department of Microbiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA.
| | - Joseph A Fraietta
- Center for Cellular Immunotherapies, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA.
- Abramson Cancer Center, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA.
- Department of Microbiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA.
- Parker Institute for Cancer Immunotherapy, University of Pennsylvania, Philadelphia, PA, USA.
- Department of Pathology and Laboratory Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA.
| |
Collapse
|
21
|
Nastoupil LJ, Andersen CR, Ayers A, Wang Y, Habermann TM, Chihara D, Kahl BS, Link BK, Koff JL, Cohen JB, Martin P, Lossos IS, Stanchina M, Haddadi S, Casulo C, Ayyappan S, Lin R, Li Z, Larson MA, Maurer MJ, Huynh L, Gao C, Ramasubramanian R, Duh MS, Mutebi A, Wang T, Jun M, Wang A, Kamalakar R, Kalsekar A, Cerhan JR, Flowers CR. Real-World Effectiveness of Chemoimmunotherapy and Novel Therapies for Patients With Relapsed/Refractory Aggressive Large B-Cell Lymphoma. CLINICAL LYMPHOMA, MYELOMA & LEUKEMIA 2025; 25:e183-e199.e8. [PMID: 39966020 DOI: 10.1016/j.clml.2024.11.014] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/13/2024] [Revised: 11/15/2024] [Accepted: 11/25/2024] [Indexed: 02/20/2025]
Abstract
INTRODUCTION Clinical trials provide meaningful data regarding the safety and efficacy of novel therapies but there is often a lag between the time of new drug approval and information on posttreatment clinical outcomes in real-world practice. This study evaluated clinical outcomes in a large real-world population of patients with relapsed and/or refractory large B-cell lymphoma (r/r LBCL) treated with chemoimmunotherapy or novel therapies in second or later lines of therapy (2L+). MATERIALS AND METHODS Data from the Lymphoma Epidemiology of Outcomes (LEO) Consortium of Real-World Evidence (CReWE) cohort (1/1/2015-2/15/2023) were analyzed. Patients' demographic and clinical characteristics were described and response rates, duration of response, progression-free survival, and overall survival were evaluated. Multivariable Cox proportional hazards regression models were used to assess associations between patient clinical characteristics and outcomes. RESULTS The 2L+ cohort included patients treated with chemoimmunotherapy (N = 593), lenalidomide-based therapy (n = 60), polatuzumab vedotin-based therapy (N = 116), tafasitamab-based therapy (N = 55), and loncastuximab tesirine (N = 42). Most patients who received prior chimeric antigen receptor T-cell therapy (CAR-T) were refractory to the treatment. Across all patients, overall response rates were <50%, with one-quarter achieving complete response and median duration of response and overall survival were short (<6 and <10 months, respectively) among patients treated with chemoimmunotherapy or novel therapies. The prognosis was worse for patients who had previously received CAR-T. Primary refractory status, high-risk disease, and failing 3 or more lines of therapy were significantly associated with worse outcomes. CONCLUSION Patients with r/r LBCL have unfavorable outcomes and need more effective treatment alternatives.
Collapse
Affiliation(s)
| | - Clark R Andersen
- Department of Lymphoma/Myeloma, MD Anderson Cancer Center, Houston, TX
| | - Amy Ayers
- Department of Lymphoma/Myeloma, MD Anderson Cancer Center, Houston, TX
| | - Yucai Wang
- Department of Hematology, Mayo Clinic, Rochester, MN
| | | | - Dai Chihara
- Department of Lymphoma/Myeloma, MD Anderson Cancer Center, Houston, TX
| | - Brad S Kahl
- Division of Oncology, Washington University School of Medicine in St. Louis, St. Louis, MO
| | - Brian K Link
- Department of Internal Medicine, University of Iowa Carver College of Medicine, Iowa City, IA
| | - Jean L Koff
- Department of Hematology and Medical Oncology, Emory University, Atlanta, GA
| | - Jonathon B Cohen
- Department of Hematology and Medical Oncology, Emory University, Atlanta, GA
| | - Peter Martin
- Division of Hematology/Oncology, Weill Medical College of Cornell Medicine, New York, NY
| | - Izidore S Lossos
- Division of Hematology, University of Miami Health System, Miami, FL
| | - Michele Stanchina
- Division of Hematology, University of Miami Health System, Miami, FL
| | - Sara Haddadi
- Department of Medicine, University of Miami Miller School of Medicine, Miami, FL
| | - Carla Casulo
- University of Rochester Medical Center, Rochester, NY
| | - Sabarish Ayyappan
- Department of Internal Medicine, University of Iowa Carver College of Medicine, Iowa City, IA
| | - Ruitao Lin
- Department of Biostatistics MD Anderson Cancer Center, Houston, TX
| | - Ziyi Li
- Department of Biostatistics MD Anderson Cancer Center, Houston, TX
| | - Melissa A Larson
- Department of Quantitative Health Sciences, Mayo Clinic, Rochester, MN
| | - Matthew J Maurer
- Department of Quantitative Health Sciences, Mayo Clinic, Rochester, MN
| | | | - Chi Gao
- Analysis Group, Inc., Boston, MA
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
22
|
Shumilov E, Levien L, Mazzeo P, Jung W, Leha A, Koch R, Hasenkamp J, Wulf G. Allogeneic stem cell transplantation against aggressive lymphomas: graft-versus-lymphoma effects in peripheral T-cell lymphoma and diffuse large B-cell lymphoma after myeloablative conditioning. Leuk Lymphoma 2025; 66:668-679. [PMID: 39660415 DOI: 10.1080/10428194.2024.2438805] [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: 07/24/2024] [Revised: 11/23/2024] [Accepted: 12/01/2024] [Indexed: 12/12/2024]
Abstract
Allogeneic stem cell transplantation (alloSCT) represents a curative option for patients with relapsed/refractory (r/r) aggressive lymphomas. We compared outcomes of alloSCT in r/r PTCL and r/r DLBCL pts (n = 150) who underwent identical myeloablative conditioning chemotherapy, GvHD prophylaxis, and relapse management. 5-year PFS and OS were significantly superior in PTCL compared to DLBCL (56% vs. 24%; 56% vs. 28%; p ≤ 0.005). A landmark analysis (day≥ +100 post-alloSCT) markedly favored outcomes in PTCL vs. DLBCL: 5-year PFS and OS of 76% vs. 30% and 76% and 35%, respectively (p ≤ 0.003). Non-relapse mortality was comparable (35% PTCL vs. 34% DLBCL, p = 0.894), whereas post-alloSCT relapse mortality was significantly higher in DLBCL (36% vs. 10%, p = 0.0007). The occurence of limited chronic GvHD did not improve outcomes in DLBCL, whereas extensive chronic GvHD was a negative risk factor for both (HR 2.09 and 2.80, p ≤ 0.006). In conclusion, we gained evidence for strong graft-versus-lymphoma activity against PTCL but not DLBCL.
Collapse
MESH Headings
- Humans
- Lymphoma, Large B-Cell, Diffuse/therapy
- Lymphoma, Large B-Cell, Diffuse/mortality
- Lymphoma, Large B-Cell, Diffuse/pathology
- Male
- Female
- Middle Aged
- Transplantation Conditioning/methods
- Transplantation Conditioning/adverse effects
- Hematopoietic Stem Cell Transplantation/adverse effects
- Hematopoietic Stem Cell Transplantation/methods
- Graft vs Host Disease/etiology
- Graft vs Host Disease/prevention & control
- Adult
- Lymphoma, T-Cell, Peripheral/therapy
- Lymphoma, T-Cell, Peripheral/mortality
- Lymphoma, T-Cell, Peripheral/pathology
- Transplantation, Homologous
- Aged
- Graft vs Tumor Effect/immunology
- Young Adult
- Treatment Outcome
- Retrospective Studies
- Adolescent
- Myeloablative Agonists/therapeutic use
Collapse
Affiliation(s)
- Evgenii Shumilov
- Department of Hematology and Medical Oncology, University Medical Center Goettingen (UMG), Goettingen, Germany
- Department of Medicine A, Hematology, Oncology and Pneumology, University Hospital Muenster (UKM), Muenster, Germany
| | - Lena Levien
- Department of Hematology and Medical Oncology, University Medical Center Goettingen (UMG), Goettingen, Germany
| | - Paolo Mazzeo
- Department of Hematology and Medical Oncology, INDIGHO Laboratory, University Medical Center Goettingen (UMG), Goettingen, Germany
| | - Wolfram Jung
- Department of Hematology and Medical Oncology, University Medical Center Goettingen (UMG), Goettingen, Germany
| | - Andreas Leha
- Department of Medical Statistics, University Medical Center Goettingen, Goettingen, Germany
| | - Raphael Koch
- Department of Hematology and Medical Oncology, University Medical Center Goettingen (UMG), Goettingen, Germany
| | - Justin Hasenkamp
- Department of Hematology and Medical Oncology, University Medical Center Goettingen (UMG), Goettingen, Germany
| | - Gerald Wulf
- Department of Hematology and Medical Oncology, University Medical Center Goettingen (UMG), Goettingen, Germany
| |
Collapse
|
23
|
Grady CB, Li Y, Maude SL, Hexner EO, Frey NV, Porter DL, Hwang WT. Inconsistent Reporting and Definitions of Time-to-Event Endpoints in CAR T Clinical Trials: A Review. Transplant Cell Ther 2025; 31:271.e1-271.e13. [PMID: 39603418 PMCID: PMC11957941 DOI: 10.1016/j.jtct.2024.11.012] [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: 08/21/2024] [Revised: 11/05/2024] [Accepted: 11/17/2024] [Indexed: 11/29/2024]
Abstract
Clinical trials evaluating chimeric antigen receptor T-cell therapy (CAR T) commonly report time-to-event (TTE) endpoints. However, definitions are not necessarily comparable across studies and variability can lead to misinterpretation of results or inappropriate comparisons across products and studies. Amid the rapidly increasing number of published CAR T trials-many of which were used for regulatory approval-this study aims to summarize the variation in the use and reporting of TTE endpoints in CAR T trials. We include CAR T trials published January 2008 to January 2023 on PubMed that reported at least one of these TTE endpoints: overall survival (OS), progression-free survival (PFS), duration of response/remission (DOR), disease-free survival, event-free survival (EFS), relapse-free survival (RFS), time to relapse, time to progression, or time to treatment failure. We abstracted and summarized endpoint definitions, including the time origin, events, competing events, and censoring. We assessed the completeness of endpoint reporting, overall and by subgroups such as study phase, publication year, and the journal's impact factor. We included 116 publications in the analysis. The most frequently reported TTEs were OS (83%,), PFS (56%), DOR (55%), and EFS (23%). Complete reporting of endpoints was poor overall: 32%, 24%, 25%, and 56% for OS, PFS, DOR, and EFS respectively. Complete reporting was lower in articles published before 2018, in lower impact factor journals, and in phase I trials. There was also a large variability in TTE definitions among those reported. For example, among 64 studies reporting DOR, 48% used the date of response as the time origin while 20% used the date of infusion, and 31% did not report a time origin. There is substantial heterogeneity and incompleteness of TTE endpoint definitions in CAR T trials that could impact the interpretation of the study results. Improving TTE reporting, by stating the time origin, event(s) of interest, competing event(s) if any, and censoring, is required to ensure valid assessment of clinical benefit and cross-trial comparison.
Collapse
Affiliation(s)
- Connor B Grady
- Department of Biostatistics, Epidemiology, and Informatics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania; Department of Epidemiology, Harvard T.H. Chan School of Public Health, Boston, Massachusetts; Abramson Cancer Center, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Yimei Li
- Department of Biostatistics, Epidemiology, and Informatics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania; Abramson Cancer Center, University of Pennsylvania, Philadelphia, Pennsylvania; Division of Oncology, Children's Hospital of Philadelphia, Philadelphia, Pennsylvania; Department of Pediatrics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania; Center for Clinical Epidemiology and Biostatistics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Shannon L Maude
- Abramson Cancer Center, University of Pennsylvania, Philadelphia, Pennsylvania; Division of Oncology, Children's Hospital of Philadelphia, Philadelphia, Pennsylvania; Department of Pediatrics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania; Center for Cellular Immunotherapies, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Elizabeth O Hexner
- Abramson Cancer Center, University of Pennsylvania, Philadelphia, Pennsylvania; Center for Cellular Immunotherapies, University of Pennsylvania, Philadelphia, Pennsylvania; Department of Medicine, Division of Hematology/Oncology, Perelman School of Medicine and the Hospital of the University of Pennsylvania, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Noelle V Frey
- Abramson Cancer Center, University of Pennsylvania, Philadelphia, Pennsylvania; Center for Cellular Immunotherapies, University of Pennsylvania, Philadelphia, Pennsylvania; Department of Medicine, Division of Hematology/Oncology, Perelman School of Medicine and the Hospital of the University of Pennsylvania, University of Pennsylvania, Philadelphia, Pennsylvania
| | - David L Porter
- Abramson Cancer Center, University of Pennsylvania, Philadelphia, Pennsylvania; Center for Cellular Immunotherapies, University of Pennsylvania, Philadelphia, Pennsylvania; Department of Medicine, Division of Hematology/Oncology, Perelman School of Medicine and the Hospital of the University of Pennsylvania, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Wei-Ting Hwang
- Department of Biostatistics, Epidemiology, and Informatics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania; Abramson Cancer Center, University of Pennsylvania, Philadelphia, Pennsylvania; Center for Clinical Epidemiology and Biostatistics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania.
| |
Collapse
|
24
|
Kim K, Chihara D. The current understanding of chimeric antigen receptor (CAR) T-cell therapy for older patients with relapsed or refractory large B-cell lymphoma. Leuk Lymphoma 2025; 66:617-627. [PMID: 39688323 DOI: 10.1080/10428194.2024.2436606] [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: 05/07/2024] [Revised: 10/29/2024] [Accepted: 11/26/2024] [Indexed: 12/18/2024]
Abstract
Chimeric antigen receptor (CAR) T-cell therapy has changed treatment landscape of relapsed or refractory (R/R) diffuse large B-cell lymphoma (DLBCL) and more older patients have been treated with curative intent for R/R disease, including patients previously deemed unfit for autologous stem-cell transplant with a broader application of CAR T-cell therapy. Due to the unique CAR T-cell-related toxicity and special attention needed in treating older patients, optimal patient selection and management of CAR T-cell therapy in older patients are becoming more critical. More data are emerging in the field; multiple approaches, such as geriatric and frailty assessment and multi-disciplinary work with geriatrics, are being studied for CAR T-cell therapy application. Studies support the safe use of CAR T-cell therapy in older patients, however, application of geriatric assessment tools and maximizing multi-disciplinary approach to tailor supportive care are critical to reduce morbidity and improve outcomes in older patients.
Collapse
MESH Headings
- Humans
- Lymphoma, Large B-Cell, Diffuse/therapy
- Lymphoma, Large B-Cell, Diffuse/pathology
- Lymphoma, Large B-Cell, Diffuse/immunology
- Lymphoma, Large B-Cell, Diffuse/mortality
- Immunotherapy, Adoptive/methods
- Immunotherapy, Adoptive/adverse effects
- Receptors, Chimeric Antigen/immunology
- Receptors, Chimeric Antigen/metabolism
- Aged
- Neoplasm Recurrence, Local/therapy
- Drug Resistance, Neoplasm
- Treatment Outcome
- Geriatric Assessment
- T-Lymphocytes/immunology
- T-Lymphocytes/metabolism
- Age Factors
- Aged, 80 and over
- Recurrence
Collapse
Affiliation(s)
- Kunhwa Kim
- Division of Cancer Medicine, MD Anderson Cancer Center, Houston, TX, USA
| | - Dai Chihara
- Department of Lymphoma-Myeloma, Division of Cancer Medicine, MD Anderson Cancer Center, Houston, TX, USA
| |
Collapse
|
25
|
Goel U, Mian A, Sauter CS. SOHO State of the Art Updates and Next Questions | Contemporary Role of Autologous Stem Cell Transplantation for the Treatment of Relapsed/Refractory Diffuse Large B-Cell Lymphoma in the Era of Cellular Therapies. CLINICAL LYMPHOMA, MYELOMA & LEUKEMIA 2025; 25:219-225. [PMID: 39214752 DOI: 10.1016/j.clml.2024.07.020] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/12/2024] [Revised: 07/28/2024] [Accepted: 07/30/2024] [Indexed: 09/04/2024]
Abstract
Since the 1990s, the standard of care for the treatment of relapsed/refractory diffuse large B-cell lymphoma (DLBCL) had been salvage chemotherapy followed by high-dose chemotherapy and autologous stem cell transplantation (HDT-ASCT) in patients with a chemotherapy-sensitive remission. However, promising results from the recent TRANSFORM and ZUMA-7 trials evaluating the efficacy of CAR T-cell therapy versus HDT-ASCT for second line relapsed/refractory DLBCL have sought to challenge this standard of care. While these studies have established a new standard for the treatment of early relapsed and primary refractory DLBCL, significant differences in the trial design between these studies and limitations with the timing of randomization during the disease course warrant a thoughtful interpretation of the results. Additionally, the financial burden and logistic challenges of CAR T-cell administration and limited access to these therapies continue to be ongoing issues. Despite the encouraging results from these trials, HDT-ASCT continues to have a role in the treatment of DLBCL, especially in disease relapsing ≥12 months after initial therapy, and in chemo sensitive disease with a good response to salvage chemotherapy. Ongoing studies evaluating novel salvage regimens for use prior to HDT-ASCT, and future studies evaluating the role of CAR T-cell therapy in chemo sensitive disease will help determine the continued role of HDT-ASCT for relapsed/refractory DLBCL.
Collapse
Affiliation(s)
- Utkarsh Goel
- Department of Internal Medicine, Cleveland Clinic, Cleveland, OH
| | - Agrima Mian
- Department of Hematology and Medical Oncology, Taussig Cancer Center, Cleveland Clinic, Cleveland, OH
| | - Craig S Sauter
- Department of Hematology and Medical Oncology, Taussig Cancer Center, Cleveland Clinic, Cleveland, OH.
| |
Collapse
|
26
|
Barbati ZR, Charli-Joseph Y. Unveiling Primary Cutaneous B-Cell Lymphomas: New Insights into Diagnosis and Treatment Strategies. Cancers (Basel) 2025; 17:1202. [PMID: 40227781 PMCID: PMC11987940 DOI: 10.3390/cancers17071202] [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: 01/20/2025] [Revised: 03/23/2025] [Accepted: 03/26/2025] [Indexed: 04/15/2025] Open
Abstract
BACKGROUND/OBJECTIVES Primary cutaneous B-cell lymphomas (PCBCL) are a rare and heterogeneous group of non-Hodgkin lymphomas that are confined to the skin at diagnosis and exhibit a tendency for cutaneous recurrence. The 5th edition of the World Health Organization and the 2022 International Consensus Classification recognize three main subtypes: primary cutaneous follicle center lymphoma (PCFCL), primary cutaneous marginal zone lymphoma/lymphoproliferative disorder (PCMZL/LPD), and primary cutaneous diffuse large B-cell lymphoma, leg type (PCDLBCL,LT). These subtypes differ in clinical behavior, histopathologic features, immunophenotype, and molecular alterations. Diagnosis and management remain challenging for clinicians. This review aims to provide a comprehensive overview of the defining features and current treatment strategies for PCBCL. METHODS This narrative review synthesizes current literature on the clinical, morphologic, immunohistochemical, and molecular characteristics of PCBCL. It also evaluates the diagnostic utility of immunohistochemistry, gene expression profiling, and molecular assays, particularly in distinguishing primary cutaneous disease from secondary cutaneous involvement by systemic lymphomas. RESULTS PCFCL arises from germinal center B-cells and must be differentiated from nodal follicular lymphoma. PCMZL/LPD is derived from post-germinal center B-cells and is often linked to chronic antigenic stimulation. Both PCFCL and PCMZL/LPD are indolent and associated with favorable outcomes. By contrast, PCDLBCL,LT is an aggressive lymphoma characterized by genetic alterations activating the NF-κB pathway, commonly including mutations to MYD88 and CD79B. Treatment strategies vary by subtype, ranging from localized therapies for indolent lymphomas to systemic chemoimmunotherapy for aggressive PCBCL. Emerging therapies, such as Bruton tyrosine kinase inhibitors and immunoregulatory agents, are being investigated for relapsed/refractory disease. CONCLUSIONS PCBCL encompass distinct clinicopathologic entities with subtype-specific diagnostic and therapeutic considerations. While current management is guided by clinical behavior, significant knowledge gaps remain regarding the molecular mechanisms underlying skin tropism, immune evasion, and disease progression. Future research could focus on improving molecular characterization and developing personalized and immune-based therapies to enhance outcomes. This review consolidates current knowledge and highlights innovations aimed at advancing the diagnosis and treatment of PCBCL in clinical practice.
Collapse
Affiliation(s)
- Zachary R. Barbati
- Department of Pathology and Laboratory Medicine, University of California San Francisco, San Francisco, CA 94107, USA;
| | - Yann Charli-Joseph
- Dermatology and Dermatopathology Private Practice, Mexico City 01090, Mexico
| |
Collapse
|
27
|
Bock TJ, Colonne CK, Fiorenza S, Turtle CJ. Outcome correlates of approved CD19-targeted CAR T cells for large B cell lymphoma. Nat Rev Clin Oncol 2025; 22:241-261. [PMID: 39966627 DOI: 10.1038/s41571-025-00992-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/27/2025] [Indexed: 02/20/2025]
Abstract
CD19-targeted chimeric antigen receptor (CAR) T cells have provided a breakthrough in the treatment of patients with relapsed and/or refractory large B cell lymphoma (LBCL). Currently, three CD19-targeted CAR T cell products are approved by the FDA and various other regulators for the treatment of patients with LBCL: axicabtagene ciloleucel, tisagenlecleucel and lisocabtagene maraleucel. Response rates following infusion of these CD19-targeted CAR T cells have been promising; however, approximately half of treated patients show relapse within 2 years. Furthermore, receiving these agents can be associated with serious toxicities, including cytokine release syndrome and immune effector cell-associated neurotoxicity syndrome. In this Review, we summarize the factors associated with the efficacy, including response and survival outcomes, and toxicity of CD19-targeted CAR T cells in pivotal clinical trials and large real-world datasets describing the outcomes of patients with LBCL who received treatment with these products.
Collapse
MESH Headings
- Humans
- Immunotherapy, Adoptive/methods
- Immunotherapy, Adoptive/adverse effects
- Antigens, CD19/immunology
- Lymphoma, Large B-Cell, Diffuse/therapy
- Lymphoma, Large B-Cell, Diffuse/immunology
- Lymphoma, Large B-Cell, Diffuse/mortality
- Receptors, Chimeric Antigen/immunology
- Receptors, Chimeric Antigen/therapeutic use
- Receptors, Antigen, T-Cell/immunology
- Receptors, Antigen, T-Cell/therapeutic use
- Treatment Outcome
- Biological Products
Collapse
Affiliation(s)
- Tamara J Bock
- School of Medical Sciences, Faculty of Medicine and Health, University of Sydney, Camperdown, New South Wales, Australia.
| | - Chanukya K Colonne
- School of Medical Sciences, Faculty of Medicine and Health, University of Sydney, Camperdown, New South Wales, Australia
| | - Salvatore Fiorenza
- School of Medical Sciences, Faculty of Medicine and Health, University of Sydney, Camperdown, New South Wales, Australia
| | - Cameron J Turtle
- Sydney Medical School, Faculty of Medicine and Health, University of Sydney, Camperdown, New South Wales, Australia
- Royal North Shore Hospital, Sydney, New South Wales, Australia
- Translational Science and Therapeutics Division, Fred Hutchinson Cancer Center, Seattle, WA, USA
| |
Collapse
|
28
|
Tix T, Alhomoud M, Shouval R, Iacoboni G, Cliff ERS, Hansen DK, Usmani SZ, Salles G, Perales MA, Cordas Dos Santos DM, Rejeski K. Non-relapse mortality with bispecific antibodies: A systematic review and meta-analysis in lymphoma and multiple myeloma. Mol Ther 2025:S1525-0016(25)00222-9. [PMID: 40170355 DOI: 10.1016/j.ymthe.2025.03.048] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2024] [Revised: 03/04/2025] [Accepted: 03/26/2025] [Indexed: 04/03/2025] Open
Abstract
Bispecific antibodies (BsAb) are associated with distinct immune-related toxicities that impact morbidity and mortality. This systematic review and meta-analysis examined non-relapse mortality (NRM) with BsAb therapy in B-cell non-Hodgkin lymphoma (NHL) and multiple myeloma (MM). A PubMed and Embase search up to October 2024 identified 29 studies (21 NHL, 8 MM) involving 2,535 patients. The overall NRM point estimate was 4.7% (95% confidence interval [CI] 3.4%-6.4%), with a median follow-up of 12.0 months. We noted no significant difference in NRM across disease entities (NHL: 4.2%, MM: 6.2%, p = 0.22). In NHL, prespecified subgroup analyses revealed increased NRM in real-world studies compared to clinical trials. For MM, an association between NRM and higher response rates and longer follow-up was noted. Meta-regression comparing BsAb and CAR-T therapies (n = 8,592) showed no significant NRM difference when accounting for key study-level confounders (p = 0.96). Overall, infections were the leading cause of NRM, accounting for 71.8% of non-relapse deaths. Of the infection-related deaths, 48% were attributed to COVID-19. In a pre-specified sensitivity analysis excluding COVID-19 fatalities, the overall NRM estimate was 3.5% (95% CI 2.6%-4.6%). Taken together, these results provide a benchmark for the estimated NRM with BsAb therapy and highlight the paramount importance of infection reporting, prevention, and mitigation.
Collapse
Affiliation(s)
- Tobias Tix
- Department of Medicine III - Hematology/Oncology, LMU University Hospital, LMU Munich, Munich, Germany
| | - Mohammad Alhomoud
- Adult Bone Marrow Transplantation Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Roni Shouval
- Adult Bone Marrow Transplantation Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA; Department of Medicine, Weill Cornell Medical College, New York, NY, USA
| | - Gloria Iacoboni
- Department of Hematology, University Hospital Vall d'Hebron, Vall d'Hebron Institute of Oncology (VHIO), Barcelona, Spain; Department of Medicine, Universitat Autònoma de Barcelona, Bellaterra, Spain
| | - Edward R Scheffer Cliff
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA; Harvard Medical School, Boston, MA, USA; Department of Clinical Haematology, Peter MacCallum Cancer Centre and Royal Melbourne Hospital, Melbourne, Australia
| | - Doris K Hansen
- Department of Blood and Marrow Transplant and Cellular Immunotherapy, Moffitt Cancer Center, Tampa, FL, USA
| | - Saad Z Usmani
- Department of Medicine, Weill Cornell Medical College, New York, NY, USA; Myeloma Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Gilles Salles
- Department of Medicine, Weill Cornell Medical College, New York, NY, USA; Lymphoma Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Miguel-Angel Perales
- Adult Bone Marrow Transplantation Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA; Department of Medicine, Weill Cornell Medical College, New York, NY, USA
| | - David M Cordas Dos Santos
- Department of Medicine III - Hematology/Oncology, LMU University Hospital, LMU Munich, Munich, Germany; Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA; Harvard Medical School, Boston, MA, USA; Broad Institute of Massachusetts Institute of Technology (MIT) and Harvard University, Cambridge, MA, USA
| | - Kai Rejeski
- Department of Medicine III - Hematology/Oncology, LMU University Hospital, LMU Munich, Munich, Germany; Adult Bone Marrow Transplantation Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA; German Cancer Consortium (DKTK), Partner Site Munich, a partnership between the DKFZ Heidelberg and LMU University Hospital, Munich, Germany.
| |
Collapse
|
29
|
Kordic A, Phillips TJ, Weiss J. The Current State of Bispecific Antibodies and T-Cell Directed Therapy in NHL. Cancers (Basel) 2025; 17:1192. [PMID: 40227768 PMCID: PMC11988123 DOI: 10.3390/cancers17071192] [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: 02/02/2025] [Revised: 03/10/2025] [Accepted: 03/25/2025] [Indexed: 04/15/2025] Open
Abstract
Relapsed/refractory non-Hodgkin lymphoma (r/r/NHL) is an aggressive disease with overall poor response rates to chemo-immunotherapy and autologous stem-cell transplant, especially in patients with diffuse large B-cell lymphoma. Major improvements in this disease space have come through the incorporation of novel immune therapies, including CD19/CD20 directed CAR-T cells and bispecific antibodies. These exciting new therapies continue to change the landscape of treatment for r/r NHL and have been incorporated in earlier lines of therapy with demonstrated efficacy and patient safety. In this review, the role of these treatments in the management of relapsed/refractory NHL is discussed in detail along with future directions of research.
Collapse
Affiliation(s)
- Austin Kordic
- City of Hope Comprehensive Cancer Center, Department of Hematology and Hematopoietic Cell Transplantation, Division of Lymphoma, Duarte, CA 91010, USA;
| | - Tycel Jovelle Phillips
- City of Hope Comprehensive Cancer Center, Department of Hematology and Hematopoietic Cell Transplantation, Division of Lymphoma, Duarte, CA 91010, USA;
| | - Jonathan Weiss
- Rogel Comprehensive Cancer Center, University of Michigan-Ann Arbor, Ann Arbor, MI 48109, USA;
| |
Collapse
|
30
|
Jean-Louis G, Cherng HJJ. Measurable Residual Disease Testing During Treatment with Bispecific Antibodies for Lymphoma. Cancers (Basel) 2025; 17:1153. [PMID: 40227652 PMCID: PMC11988116 DOI: 10.3390/cancers17071153] [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: 02/13/2025] [Revised: 03/20/2025] [Accepted: 03/27/2025] [Indexed: 04/15/2025] Open
Abstract
The introduction of bispecific antibodies (BsAbs) has led to significant improvements in survival for patients with relapsed and refractory B-cell lymphomas. Despite these advances, there remains a significant number of patients who experience disease progression after these novel therapies. Predicting which patients may respond to certain treatments and the durability of their responses remains challenging. Measurable residual disease (MRD) has become easier to detect and quantify through the use of genomic next-generation sequencing tools and has been studied as a possible biomarker to predict long-term outcomes and risk-stratify patients after BsAb therapy in several lymphoma subtypes. Here, we review recent data demonstrating that MRD negativity is associated with radiographic response and improved progression-free survival. Because of heterogeneity in assay choice, assessment timing, and technical parameters, further work is needed before MRD testing is ready to be incorporated into clinical practice in the context of BsAb treatment for B-cell lymphomas.
Collapse
Affiliation(s)
| | - Hua-Jay J. Cherng
- Division of Hematology/Oncology, Columbia University Irving Medical Center, New York, NY 10032, USA;
| |
Collapse
|
31
|
Porazzi P, Nason S, Yang Z, Carturan A, Ghilardi G, Guruprasad P, Patel RP, Tan M, Padmanabhan AA, Lemoine J, Fardella E, Zhang Y, Pajarillo R, Chen L, Ugwuanyi O, Markowitz K, Delman D, Angelos MG, Shestova O, Isshiki Y, Blanchard T, Béguelin W, Melnick AM, Linette GP, Beatty GL, Carreno BM, Cohen IJ, Paruzzo L, Schuster SJ, Ruella M. EZH1/EZH2 inhibition enhances adoptive T cell immunotherapy against multiple cancer models. Cancer Cell 2025; 43:537-551.e7. [PMID: 39983725 DOI: 10.1016/j.ccell.2025.01.013] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/26/2024] [Revised: 11/21/2024] [Accepted: 01/29/2025] [Indexed: 02/23/2025]
Abstract
Tumor resistance to chimeric antigen receptor T cell (CAR-T) and, in general, to adoptive cell immunotherapies (ACTs) is a major challenge in the clinic. We hypothesized that inhibiting the tumor drivers' methyltransferases EZH2 and EZH1 could enhance ACT by rewiring cancer cells to a more immunogenic state. In human B cell lymphoma, EZH2 inhibition (tazemetostat) improved the efficacy of anti-CD19 CAR-T by enhancing activation, expansion, and tumor infiltration. Mechanistically, tazemetostat-treated tumors showed upregulation of genes related to adhesion, B cell activation, and inflammatory responses, and increased avidity to CAR-T. Furthermore, tazemetostat improved CAR- and TCR-engineered T cell efficacy in multiple liquid (myeloma and acute myeloid leukemia) and solid (sarcoma, ovarian, and prostate) cancers. Lastly, combined EZH1/EZH2 inhibition (valemetostat) further boosted CAR-T efficacy and expansion in multiple cancers. This study shows that EZH1/2 inhibition reprograms tumors to a more immunogenic state and potentiates ACT in preclinical models of both liquid and solid cancers.
Collapse
Affiliation(s)
- Patrizia Porazzi
- Center for Cellular Immunotherapies, University of Pennsylvania, Philadelphia, PA, USA; Division of Hematology-Oncology, Hospital of the University of Pennsylvania, Philadelphia, PA, USA; Lymphoma Program, Abramson Cancer Center, University of Pennsylvania, Philadelphia, PA, USA
| | - Siena Nason
- Center for Cellular Immunotherapies, University of Pennsylvania, Philadelphia, PA, USA; Division of Hematology-Oncology, Hospital of the University of Pennsylvania, Philadelphia, PA, USA; Lymphoma Program, Abramson Cancer Center, University of Pennsylvania, Philadelphia, PA, USA
| | - Ziqi Yang
- Center for Cellular Immunotherapies, University of Pennsylvania, Philadelphia, PA, USA; Division of Hematology-Oncology, Hospital of the University of Pennsylvania, Philadelphia, PA, USA; Lymphoma Program, Abramson Cancer Center, University of Pennsylvania, Philadelphia, PA, USA
| | - Alberto Carturan
- Center for Cellular Immunotherapies, University of Pennsylvania, Philadelphia, PA, USA; Division of Hematology-Oncology, Hospital of the University of Pennsylvania, Philadelphia, PA, USA; Lymphoma Program, Abramson Cancer Center, University of Pennsylvania, Philadelphia, PA, USA
| | - Guido Ghilardi
- Center for Cellular Immunotherapies, University of Pennsylvania, Philadelphia, PA, USA; Division of Hematology-Oncology, Hospital of the University of Pennsylvania, Philadelphia, PA, USA; Lymphoma Program, Abramson Cancer Center, University of Pennsylvania, Philadelphia, PA, USA
| | - Puneeth Guruprasad
- Center for Cellular Immunotherapies, University of Pennsylvania, Philadelphia, PA, USA; Division of Hematology-Oncology, Hospital of the University of Pennsylvania, Philadelphia, PA, USA; Lymphoma Program, Abramson Cancer Center, University of Pennsylvania, Philadelphia, PA, USA
| | - Ruchi P Patel
- Center for Cellular Immunotherapies, University of Pennsylvania, Philadelphia, PA, USA; Division of Hematology-Oncology, Hospital of the University of Pennsylvania, Philadelphia, PA, USA; Lymphoma Program, Abramson Cancer Center, University of Pennsylvania, Philadelphia, PA, USA
| | - Melody Tan
- Center for Cellular Immunotherapies, University of Pennsylvania, Philadelphia, PA, USA; Division of Hematology-Oncology, Hospital of the University of Pennsylvania, Philadelphia, PA, USA; Lymphoma Program, Abramson Cancer Center, University of Pennsylvania, Philadelphia, PA, USA; Parker Institute for Cancer Immunotherapy, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Anushka Anant Padmanabhan
- Center for Cellular Immunotherapies, University of Pennsylvania, Philadelphia, PA, USA; Division of Hematology-Oncology, Hospital of the University of Pennsylvania, Philadelphia, PA, USA; Lymphoma Program, Abramson Cancer Center, University of Pennsylvania, Philadelphia, PA, USA
| | - Jean Lemoine
- Center for Cellular Immunotherapies, University of Pennsylvania, Philadelphia, PA, USA; Division of Hematology-Oncology, Hospital of the University of Pennsylvania, Philadelphia, PA, USA; Lymphoma Program, Abramson Cancer Center, University of Pennsylvania, Philadelphia, PA, USA
| | - Eugenio Fardella
- Center for Cellular Immunotherapies, University of Pennsylvania, Philadelphia, PA, USA; Division of Hematology-Oncology, Hospital of the University of Pennsylvania, Philadelphia, PA, USA; School of Medicine, Università degli Studi di Milano, Milan, Italy
| | - Yunlin Zhang
- Center for Cellular Immunotherapies, University of Pennsylvania, Philadelphia, PA, USA; Division of Hematology-Oncology, Hospital of the University of Pennsylvania, Philadelphia, PA, USA; Lymphoma Program, Abramson Cancer Center, University of Pennsylvania, Philadelphia, PA, USA
| | - Raymone Pajarillo
- Center for Cellular Immunotherapies, University of Pennsylvania, Philadelphia, PA, USA; Division of Hematology-Oncology, Hospital of the University of Pennsylvania, Philadelphia, PA, USA; Lymphoma Program, Abramson Cancer Center, University of Pennsylvania, Philadelphia, PA, USA
| | - Linhui Chen
- Center for Cellular Immunotherapies, University of Pennsylvania, Philadelphia, PA, USA; Division of Hematology-Oncology, Hospital of the University of Pennsylvania, Philadelphia, PA, USA; Lymphoma Program, Abramson Cancer Center, University of Pennsylvania, Philadelphia, PA, USA
| | - Ositadimma Ugwuanyi
- Center for Cellular Immunotherapies, University of Pennsylvania, Philadelphia, PA, USA; Division of Hematology-Oncology, Hospital of the University of Pennsylvania, Philadelphia, PA, USA; Lymphoma Program, Abramson Cancer Center, University of Pennsylvania, Philadelphia, PA, USA
| | - Kelly Markowitz
- Division of Hematology-Oncology, Hospital of the University of Pennsylvania, Philadelphia, PA, USA
| | - Devora Delman
- Division of Hematology-Oncology, Hospital of the University of Pennsylvania, Philadelphia, PA, USA
| | - Mathew G Angelos
- Center for Cellular Immunotherapies, University of Pennsylvania, Philadelphia, PA, USA; Division of Hematology-Oncology, Hospital of the University of Pennsylvania, Philadelphia, PA, USA; Lymphoma Program, Abramson Cancer Center, University of Pennsylvania, Philadelphia, PA, USA
| | - Olga Shestova
- Center for Cellular Immunotherapies, University of Pennsylvania, Philadelphia, PA, USA; Division of Hematology-Oncology, Hospital of the University of Pennsylvania, Philadelphia, PA, USA
| | - Yusuke Isshiki
- Division of Hematology/Oncology, Department of Medicine, Weill Cornell Medicine, Cornell University, New York, NY, USA
| | - Tatiana Blanchard
- Center for Cellular Immunotherapies, University of Pennsylvania, Philadelphia, PA, USA
| | - Wendy Béguelin
- Division of Hematology/Oncology, Department of Medicine, Weill Cornell Medicine, Cornell University, New York, NY, USA
| | - Ari M Melnick
- Division of Hematology/Oncology, Department of Medicine, Weill Cornell Medicine, Cornell University, New York, NY, USA
| | - Gerald P Linette
- Center for Cellular Immunotherapies, University of Pennsylvania, Philadelphia, PA, USA; Parker Institute for Cancer Immunotherapy, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Gregory L Beatty
- Division of Hematology-Oncology, Hospital of the University of Pennsylvania, Philadelphia, PA, USA; Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Beatriz M Carreno
- Center for Cellular Immunotherapies, University of Pennsylvania, Philadelphia, PA, USA; Parker Institute for Cancer Immunotherapy, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Ivan J Cohen
- Center for Cellular Immunotherapies, University of Pennsylvania, Philadelphia, PA, USA; Division of Hematology-Oncology, Hospital of the University of Pennsylvania, Philadelphia, PA, USA; Lymphoma Program, Abramson Cancer Center, University of Pennsylvania, Philadelphia, PA, USA
| | - Luca Paruzzo
- Center for Cellular Immunotherapies, University of Pennsylvania, Philadelphia, PA, USA; Division of Hematology-Oncology, Hospital of the University of Pennsylvania, Philadelphia, PA, USA; Lymphoma Program, Abramson Cancer Center, University of Pennsylvania, Philadelphia, PA, USA
| | - Stephen J Schuster
- Center for Cellular Immunotherapies, University of Pennsylvania, Philadelphia, PA, USA; Division of Hematology-Oncology, Hospital of the University of Pennsylvania, Philadelphia, PA, USA; Lymphoma Program, Abramson Cancer Center, University of Pennsylvania, Philadelphia, PA, USA
| | - Marco Ruella
- Center for Cellular Immunotherapies, University of Pennsylvania, Philadelphia, PA, USA; Division of Hematology-Oncology, Hospital of the University of Pennsylvania, Philadelphia, PA, USA; Lymphoma Program, Abramson Cancer Center, University of Pennsylvania, Philadelphia, PA, USA.
| |
Collapse
|
32
|
Carter S, Wigmore T. Immunotherapy on ICU: a narrative review. Anaesthesia 2025; 80:299-310. [PMID: 39776055 DOI: 10.1111/anae.16453] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/17/2024] [Indexed: 01/11/2025]
Abstract
BACKGROUND Patients with cancer account for 15% of all admissions to critical care and so an understanding of the pathophysiology and anticipated complications of specialist treatment is essential for the intensive care clinician. The development of chimeric antigen receptor T-cell therapy for haematological malignancies and immune checkpoint inhibitors for solid organ tumours has led to significant improvements in the prognosis of those patients whose tumours respond. This review is intended to provide the non-specialist with an understanding of the current concepts in pathophysiology, diagnosis and management of complications due to chimeric antigen receptor T-cell therapy and immune checkpoint inhibitors for malignant disease. METHODS We performed searches of electronic databases to identify relevant peer-reviewed publications in the literature. Basic science; clinical trials; cohort studies; systematic reviews; meta-analyses; and guidelines were eligible for inclusion. Abstracts were screened to identify publications relevant to immune effector cell toxicities of chimeric antigen receptor T-cell therapy and immune-related adverse events of immune checkpoint inhibitors. RESULTS While the pathophysiology for toxicities due to chimeric antigen receptor T-cells and immune checkpoint inhibitors remains incompletely understood, targeted drug therapies have been successfully implemented for toxicities such as cytokine release syndrome. Corticosteroids remain an important component of pharmacological management. The diagnosis of toxicities remains largely clinical, and a high index of suspicion should remain for infective complications. Management of toxicities should be undertaken in conjunction with the patient's primary oncologist. CONCLUSION Despite significant advances in the development of targeted immunotherapy, the mechanism of action for the resultant toxicities remains poorly understood and limits the development of predictive models, diagnostic biomarkers and highly effective treatment options. Further research is needed to identify treatment regimens which minimise the use of corticosteroids in chimeric antigen receptor T-cell and immune checkpoint inhibitor-associated toxicities.
Collapse
Affiliation(s)
- Sef Carter
- East of England School of Anaesthesia, Basildon, UK
| | - Timothy Wigmore
- Department of Critical Care, The Royal Marsden Hospital, London, UK
| |
Collapse
|
33
|
Yamauchi N, Maruyama D. Current treatment approach and future perspectives in B cell lymphoma. Int J Hematol 2025; 121:342-355. [PMID: 39572467 DOI: 10.1007/s12185-024-03879-w] [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: 10/01/2024] [Revised: 11/04/2024] [Accepted: 11/10/2024] [Indexed: 02/26/2025]
Abstract
Diffuse large B cell lymphoma (DLBCL) and follicular lymphoma (FL) represent the two major subtypes of mature B cell lymphoma. A deeper understanding of tumor biology, as well as molecular classification characterized by targetable gene alterations, and the introduction of novel treatment options, including targeted drugs (e.g., antibody-drug conjugates and small molecules [e.g., Bruton tyrosine kinase inhibitor]) and immune therapies (e.g., chimeric antigen receptor [CAR] T cell therapy and bispecific antibody [BsAb]), has changed the treatment paradigms for DLBCL and FL. In clinical practice, however, treatment regimens are determined mainly based on prior treatment history, duration of response after previous treatment, patient age, and patient frailty because there have been few randomized trials to inform treatment selection for patients with relapsed or refractory disease and because there is no single prognostic index that guides suitable treatment for each patient. In this review, we summarize the treatment options for DLBCL and FL and discuss the treatment strategies for these two subtypes. We also discuss future perspectives for the treatment of these subtypes.
Collapse
Affiliation(s)
- Nobuhiko Yamauchi
- Department of Hematology Oncology, Cancer Institute Hospital, Japanese Foundation for Cancer Research, 3-8-31, Ariake, Koto-Ku, Tokyo, 135-8550, Japan
| | - Dai Maruyama
- Department of Hematology Oncology, Cancer Institute Hospital, Japanese Foundation for Cancer Research, 3-8-31, Ariake, Koto-Ku, Tokyo, 135-8550, Japan.
| |
Collapse
|
34
|
Dickinson M, O'Leary N, Hamad N, Cochrane T, Ho PJ, Cheah CY, Bishop D, Gregory GP, Butler J, Barraclough A. Establishing best practice in the Australian haematology setting for the use of chimeric antigen receptor T-cell therapy for relapsed and refractory lymphoma. Intern Med J 2025; 55 Suppl 2:4-27. [PMID: 40171767 DOI: 10.1111/imj.16544] [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: 02/13/2024] [Accepted: 09/24/2024] [Indexed: 04/04/2025]
Abstract
Autologous CD19 chimeric antigen receptor T (CAR T)-cell therapies have significantly improved clinical outcomes for many patients with relapsed and refractory (R/R) lymphoma. However, the process of delivering CAR T-cell therapy is specialised and complex, in part due to specific post-infusion toxicities. Several CAR T-cell products are now available in Australia, although not all states have treatment centres. In this review, we aim to define best practice for the referral and treatment of patients with R/R B-cell lymphoma with CAR T-cell therapy in Australia. We outline the processes for referral, optimal patient selection and best practice in the management of patients receiving CAR T cells.
Collapse
Affiliation(s)
- Michael Dickinson
- Peter MacCallum Cancer Centre, Royal Melbourne Hospital and the Sir Peter MacCallum Department of Oncology at the University of Melbourne, Sydney, New South Wales, Australia
| | - Nicole O'Leary
- Peter MacCallum Cancer Centre, Royal Melbourne Hospital and the Sir Peter MacCallum Department of Oncology at the University of Melbourne, Sydney, New South Wales, Australia
| | - Nada Hamad
- Department of Haematology, St Vincent's Hospital, Sydney, New South Wales, Australia
- School of Clinical Medicine, Faculty of Medicine and Health, University of NSW, Sydney, New South Wales, Australia
- School of Medicine, University of Notre Dame, Sydney, New South Wales, Australia
| | - Tara Cochrane
- Department of Haematology, Gold Coast University Hospital, Southport, Queensland, Australia
- School of Medicine and Dentistry, Griffith University, Southport, Queensland, Australia
| | - P Joy Ho
- Department of Haematology, Royal Prince Alfred Hospital and University of Sydney, Sydney, New South Wales, Australia
| | - Chan Y Cheah
- Department of Haematology, Sir Charles Gairdner Hospital, Nedlands, and University of Western Australia, Perth, Western Australia, Australia
| | - David Bishop
- Department of Haematology, Westmead Hospital, Sydney, New South Wales, Australia
- Sydney Medical School, Faculty of Medicine and Health, The University of Sydney, Sydney, New South Wales, Australia
| | - Gareth P Gregory
- School of Clinical Sciences at Monash Health, Monash University, Melbourne, Victoria, Queensland, Australia
| | - Jason Butler
- Department of Bone Marrow Transplantation, Royal Brisbane and Women's Hospital, Brisbane, Queensland, Australia
| | - Allison Barraclough
- Department of Haematology, Fiona Stanley Hospital, Perth, Western Australia, Australia
| |
Collapse
|
35
|
Ennishi D. Recent advances in understanding of pathogenesis and treatment development for diffuse large B-cell lymphoma and follicular lymphoma. Int J Hematol 2025; 121:318-320. [PMID: 39928216 DOI: 10.1007/s12185-025-03939-9] [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: 01/19/2025] [Revised: 01/19/2025] [Accepted: 01/23/2025] [Indexed: 02/11/2025]
Abstract
Diffuse large B-cell lymphoma (DLBCL) and follicular lymphoma (FL) are the most common histological types of B-cell lymphoma, but have significantly different pathological and biological characteristics. In recent years, understanding of the genetic abnormalities and microenvironmental structures of these lymphomas has progressed, and various molecular targeted drugs and immunotherapies have been introduced into clinical practice. Therefore, accurate understanding of etiology and its clinical relevance is required for the appropriate management of these lymphomas.
Collapse
MESH Headings
- Humans
- Lymphoma, Large B-Cell, Diffuse/therapy
- Lymphoma, Large B-Cell, Diffuse/etiology
- Lymphoma, Large B-Cell, Diffuse/genetics
- Lymphoma, Large B-Cell, Diffuse/pathology
- Lymphoma, Follicular/therapy
- Lymphoma, Follicular/etiology
- Lymphoma, Follicular/genetics
- Lymphoma, Follicular/pathology
- Tumor Microenvironment
- Molecular Targeted Therapy
- Immunotherapy
Collapse
Affiliation(s)
- Daisuke Ennishi
- Center for Comprehensive Genomic Medicine, Okayama University Hospital, Okayama, Japan.
| |
Collapse
|
36
|
Gao X, Lyu T, Xu M, Hampson LV, Du Y, Lin R, Yateman N, Tian L, Sun J. RMST for Interval-Censored Data in Oncology Clinical Trials. Stat Med 2025; 44:e70012. [PMID: 39921572 DOI: 10.1002/sim.70012] [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: 01/03/2024] [Revised: 11/19/2024] [Accepted: 01/18/2025] [Indexed: 02/10/2025]
Abstract
In oncology studies, the assumption of proportional hazards is often questionable due to factors such as the presence of cured patients, a delayed treatment benefit, and possible treatment switching. The restricted mean survival time (RMST) has emerged as a valuable alternative summary measure to the hazard ratio (HR) in this scenario as it provides a clinically meaningful interpretation of treatment benefit without additional assumptions. As a commonly used primary endpoint, progression-free survival (PFS) is defined as the time from randomization to the first occurrence of death or progression of disease (PD). However, PFS involves dual observation processes where, in practice, the exact death time is typically recorded, but PD is interval-censored. This feature is also present in other commonly used primary endpoints, including event-free survival, disease-free survival, and relapse-free survival. The conventional approach imputes the PD time with the right boundary of the time interval during which the PD occurs. This paper presents alternative estimation and inference approaches to estimate RMST with a mixture of right-censored and interval-censored data. Different approaches are explored by simulation under various plausible scenarios for oncology clinical trials with regard to the assessment frequency, randomness in the actual assessment times, and size of treatment effect. The choice of the restricted time point in RMST is also explored. The simulation results indicate that the RMST estimators that take account of the interval censoring inherent in the data are unbiased and more accurate than the conventional estimators, while the performance for two-group comparisons is comparable. Furthermore, the performance of the proposed estimators is contingent on the scheduled assessment plan and patients' visit window.
Collapse
Affiliation(s)
- Xiyuan Gao
- Department of Statistics, University of Missouri-Columbia, Columbia, Missouri, USA
| | - Tianmeng Lyu
- Novartis Pharmaceuticals Corporation, East Hanover, New Jersey, USA
| | - Menghao Xu
- School of Statistics, East China Normal University, Shanghai, China
| | | | - Yan Du
- Novartis Institutes for Biomedical Research Co., Shanghai, China
| | - Renxin Lin
- Novartis Institutes for Biomedical Research Co., Shanghai, China
| | | | - Lu Tian
- Department of Biomedical Data Science, Stanford University, Stanford, California, USA
| | - Jianguo Sun
- Department of Statistics, University of Missouri-Columbia, Columbia, Missouri, USA
| |
Collapse
|
37
|
Liaskas A, Dimopoulou MN, Piperidou A, Angelopoulou MK, Vassilakopoulos TP. Current Issues and Future Perspectives of Targeted Therapies in Primary Mediastinal Large B-Cell Lymphoma. J Clin Med 2025; 14:1191. [PMID: 40004722 PMCID: PMC11856677 DOI: 10.3390/jcm14041191] [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: 01/02/2025] [Revised: 01/31/2025] [Accepted: 02/04/2025] [Indexed: 02/27/2025] Open
Abstract
Primary mediastinal large B-cell lymphoma (PMLBCL) is a rare, aggressive B-cell lymphoma, sharing common features with diffuse large B-cell lymphoma (DLBCL) and Hodgkin lymphoma (HL). PMLBCL is usually cured with single-hit immunochemotherapy in the first-line setting. Relapses tend to be aggressive and may be unresponsive to conventional chemotherapy. Autologous stem cell transplant (ASCT) remains a viable option for chemosensitive patients; nevertheless, targeted therapies appear to be highly promising. Checkpoint inhibitors (CPIs) have already transformed the course of relapse/refractory disease, while CD-19-directed Chimeric Antigen Receptor (CAR) T-cell therapy may produce remarkably favorable outcomes. The exact position of CAR T-cells and CPIs in the treatment algorithm, along with the role of radiotherapy and ASCT, remains to be precisely determined. In the current review, we aim to present the recent research on targeted agents in PMLBCL and define their sequencing within the treatment algorithm, mainly in the relapse/refractory setting.
Collapse
Affiliation(s)
| | - Maria N. Dimopoulou
- Department of Hematology and Bone Marrow Transplantation, Medical School, National and Kapodistrian University of Athens, General Hospital of Athens “Laikon”, 11527 Athens, Greece; (A.L.); (A.P.); (M.K.A.); (T.P.V.)
| | | | | | | |
Collapse
|
38
|
Thiruvengadam SK, Merryman R, Wang Y, Gaulin C, Bezerra E, Voorhees T, Seshadri MR, Falade A, Habib A, Ayers AA, Bailey M, Brown A, Bailey N, Patel K, Andreadis CB, Kittai AS, Jacobson C, Palmer J, Forman SJ, Nastoupil L, Budde LE. Outcomes of CD19 CAR T in Transformed Indolent Lymphoma Compared to De Novo Aggressive Large B-Cell Lymphoma. Am J Hematol 2025; 100:236-248. [PMID: 39715004 PMCID: PMC11705210 DOI: 10.1002/ajh.27548] [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: 08/13/2024] [Revised: 11/12/2024] [Accepted: 11/22/2024] [Indexed: 12/25/2024]
Abstract
Chimeric antigen receptor (CAR) T-cell therapy has revolutionized treatment of aggressive large B-cell lymphoma (aLBCL). Patients with transformed indolent non-Hodgkin lymphoma (tiNHL) were included in key CAR trials, but outcomes of CAR for this distinct, historically high-risk group are poorly understood. We conducted a multicenter retrospective study of 1182 patients with aLBCL receiving standard-of-care CAR T between 2017 and 2022, including 338 (29%) with tiNHL. Rates of grade ≥ 3 cytokine release syndrome (CRS) were similar between tiNHL and de novo cohorts (7% vs. 8%, p = 0.6), while grade ≥ 3 immune effector cell-associated neurotoxicity syndrome was lower in tiNHL (21% vs. 27%, p = 0.02). Overall response rate was similar in both cohorts (83% vs. 81%, p = 0.3), while complete response rate was higher in tiNHL (67% vs. 59%, p = 0.017). With a median follow-up of 22.3 months, the progression/relapse-free (PFS) and overall survival (OS) were similar between the tiNHL and de novo cohorts (24-month PFS 41% [95% CI: 35%-46%] vs. 38% [95% CI: 35%-42%]; 24-month OS 58% [95% CI: 52%-63%] vs. 52% [95% CI: 48%-56%], respectively). After adjusting for key risk factors, there was a trend toward a lower hazard of disease progression, relapse or death post-CAR for tiNHL patients compared to de novo aLBCL patients (HR: 0.84 [95% CI: 0.69-1.0], p = 0.07). Elevated LDH, advanced stage, prior bendamustine within 12 months of CAR, receipt of bridging therapy, CNS involvement, and ≥ 3 prior lines of therapy were each associated with inferior PFS. In conclusion, CAR T therapy is highly effective with an acceptable toxicity profile in patients with tiNHL.
Collapse
Affiliation(s)
| | - Reid Merryman
- Department of Hematologic OncologyDana Farber Cancer InstituteBostonMassachusettsUSA
| | - Yan Wang
- Department of Computational and Quantitative Medicine, Division of BiostatisticsCity of Hope National Medical CenterDuarteCaliforniaUSA
| | - Charles Gaulin
- Department of Lymphoma‐Myeloma, Division of Cancer MedicineMD Anderson Cancer CenterHoustonTexasUSA
| | - Evandro Bezerra
- Division of HematologyThe Ohio State UniversityColumbusOhioUSA
| | | | - Madhav R. Seshadri
- Division of Hematology and OncologyUniversity of California San FranciscoSan FranciscoCaliforniaUSA
| | - Ayo Falade
- Department of MedicineMass General Brigham Salem HospitalSalemNew HampshireUSA
| | - Alma Habib
- Division of HematologyThe Ohio State UniversityColumbusOhioUSA
| | - Amy A. Ayers
- Department of Lymphoma‐Myeloma, Division of Cancer MedicineMD Anderson Cancer CenterHoustonTexasUSA
| | - Megumi Bailey
- Center for Blood Disorders and Cellular TherapySwedish Cancer InstituteSeattleWashingtonUSA
| | - Annette Brown
- Department of Hematology and Hematopoietic Cell TransplantationCity of Hope National Medical CenterDuarteCaliforniaUSA
| | - Neil Bailey
- Center for Blood Disorders and Cellular TherapySwedish Cancer InstituteSeattleWashingtonUSA
| | - Krish Patel
- Center for Blood Disorders and Cellular TherapySwedish Cancer InstituteSeattleWashingtonUSA
| | - Charalambos B. Andreadis
- Division of Hematology and OncologyUniversity of California San FranciscoSan FranciscoCaliforniaUSA
| | - Adam S. Kittai
- Department of Hematology and Medical OncologyIcahn School of Medicine at Mount SinaiNew YorkNew YorkUSA
| | - Caron Jacobson
- Department of Hematologic OncologyDana Farber Cancer InstituteBostonMassachusettsUSA
| | - Joycelynne Palmer
- Department of Hematology and Hematopoietic Cell TransplantationCity of Hope National Medical CenterDuarteCaliforniaUSA
- Department of Computational and Quantitative Medicine, Division of BiostatisticsCity of Hope National Medical CenterDuarteCaliforniaUSA
| | - Stephen J. Forman
- Department of Hematology and Hematopoietic Cell TransplantationCity of Hope National Medical CenterDuarteCaliforniaUSA
| | - Loretta Nastoupil
- Department of Lymphoma‐Myeloma, Division of Cancer MedicineMD Anderson Cancer CenterHoustonTexasUSA
| | - Lihua E. Budde
- Department of Hematology and Hematopoietic Cell TransplantationCity of Hope National Medical CenterDuarteCaliforniaUSA
| |
Collapse
|
39
|
Panuccio G, Correale P, d'Apolito M, Mutti L, Giannicola R, Pirtoli L, Giordano A, Labate D, Macheda S, Carabetta N, Abdelwahed YS, Landmesser U, Tassone P, Tagliaferri P, De Rosa S, Torella D. Immuno-related cardio-vascular adverse events associated with immuno-oncological treatments: an under-estimated threat for cancer patients. Basic Res Cardiol 2025; 120:153-169. [PMID: 39225869 PMCID: PMC11790807 DOI: 10.1007/s00395-024-01077-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/22/2024] [Revised: 08/16/2024] [Accepted: 08/17/2024] [Indexed: 09/04/2024]
Abstract
Immunotherapy represents an emergent and heterogeneous group of anticancer treatments harnessing the human immune-surveillance system, including immune-checkpoint inhibitor monoclonal antibodies (mAbs), Chimeric Antigen Receptor T Cells (CAR-T) therapy, cancer vaccines and lymphocyte activation gene-3 (LAG-3) therapy. While remarkably effective against several malignancies, these therapies, often in combination with other cancer treatments, have showed unforeseen toxicity, including cardiovascular complications. The occurrence of immuno-mediated adverse (irAEs) events has been progressively reported in the last 10 years. These irAEs present an extended range of severity, from self-limiting to life-threatening conditions. Although recent guidelines in CardioOncology have provided important evidence in managing cancer treatments, they often encompass general approaches. However, a specific focus is required due to the particular etiology, unique risk factors, and associated side effects of immunotherapy. This review aims to deepen the understanding of the prevalence and nature of cardiovascular issues in patients undergoing immunotherapy, offering insights into strategies for risk stratification and management.
Collapse
Affiliation(s)
- Giuseppe Panuccio
- Department of Cardiology, Angiology and Intensive Care Medicine, Deutsches Herzzentrum der Charité Berlin, 12200, Berlin, Germany.
- Department of Experimental and Clinical Medicine, Magna Graecia University, Catanzaro, Italy.
| | - Pierpaolo Correale
- Medical Oncology Unit, Grande Ospedale Metropolitano Bianchi Melacrino Morelli, 89124, Reggio Calabria, Italy
- Sbarro Institute for Cancer Research and Molecular Medicine and Center of Biotechnology, College of Science and Technology, Temple University, Philadelphia, PA, 19122, USA
| | - Maria d'Apolito
- Department of Experimental and Clinical Medicine, Magna Graecia University, Catanzaro, Italy
- Medical Oncology Unit, Grande Ospedale Metropolitano Bianchi Melacrino Morelli, 89124, Reggio Calabria, Italy
| | - Luciano Mutti
- Sbarro Institute for Cancer Research and Molecular Medicine and Center of Biotechnology, College of Science and Technology, Temple University, Philadelphia, PA, 19122, USA
- Department of Applied Sciences and Biotechnology, Università dell'Aquila, L'Aquila, Italy
| | - Rocco Giannicola
- Medical Oncology Unit, Grande Ospedale Metropolitano Bianchi Melacrino Morelli, 89124, Reggio Calabria, Italy
| | - Luigi Pirtoli
- Sbarro Institute for Cancer Research and Molecular Medicine and Center of Biotechnology, College of Science and Technology, Temple University, Philadelphia, PA, 19122, USA
| | - Antonio Giordano
- Sbarro Institute for Cancer Research and Molecular Medicine and Center of Biotechnology, College of Science and Technology, Temple University, Philadelphia, PA, 19122, USA
- Department of Medical Biotechnology, University of Siena, 53100, Siena, Italy
| | - Demetrio Labate
- Unit of Intensive Care Medicine and Anesthesia, Grande Ospedale Metropolitano Bianchi Melacrino Morelli, 89124, Reggio Calabria, Italy
| | - Sebastiano Macheda
- Unit of Intensive Care Medicine and Anesthesia, Grande Ospedale Metropolitano Bianchi Melacrino Morelli, 89124, Reggio Calabria, Italy
| | - Nicole Carabetta
- Department of Medical and Surgical Sciences, Magna Graecia University, Catanzaro, Italy
| | - Youssef S Abdelwahed
- Department of Cardiology, Angiology and Intensive Care Medicine, Deutsches Herzzentrum der Charité Berlin, 12200, Berlin, Germany
- DZHK (German Centre for Cardiovascular Research), 10785, Berlin, Germany
| | - Ulf Landmesser
- Department of Cardiology, Angiology and Intensive Care Medicine, Deutsches Herzzentrum der Charité Berlin, 12200, Berlin, Germany
- DZHK (German Centre for Cardiovascular Research), 10785, Berlin, Germany
- Berlin Institute of Health (BIH), 10178, Berlin, Germany
| | - Pierfrancesco Tassone
- Department of Experimental and Clinical Medicine, Magna Graecia University, Catanzaro, Italy
- Sbarro Institute for Cancer Research and Molecular Medicine and Center of Biotechnology, College of Science and Technology, Temple University, Philadelphia, PA, 19122, USA
| | - Pierosandro Tagliaferri
- Department of Experimental and Clinical Medicine, Magna Graecia University, Catanzaro, Italy
| | - Salvatore De Rosa
- Department of Medical and Surgical Sciences, Magna Graecia University, Catanzaro, Italy
| | - Daniele Torella
- Department of Experimental and Clinical Medicine, Magna Graecia University, Catanzaro, Italy.
| |
Collapse
|
40
|
van den Berg J, Läubli H, Khanna N, Jeker LT, Holbro A. Basic Concepts and Indications of CAR T Cells. Hamostaseologie 2025; 45:14-23. [PMID: 39970899 DOI: 10.1055/a-2491-3652] [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: 02/21/2025] Open
Abstract
Chimeric antigen receptor (CAR) T cell therapy has revolutionized cancer immunotherapy, particularly for hematological malignancies. This personalized approach is based on genetically engineering T cells derived from the patient to target antigens expressed-among others-on malignant cells. Nowadays they offer new hope where conventional therapies, such as chemotherapy and radiation, have often failed. Since the first FDA approval in 2017, CAR T cell therapy has rapidly expanded, proving highly effective against previously refractory diseases with otherwise a dismal outcome. Despite its promise, CAR T cell therapy continues to face significant challenges, including complex manufacturing, the management of toxicities, resistance mechanisms that impact long-term efficacy, and limited access as well as high costs, which continue to shape ongoing research and clinical applications. This review aims to provide an overview of CAR T cell therapy, including its fundamental concepts, clinical applications, current challenges, and future directions in hematological malignancies.
Collapse
Affiliation(s)
- Jana van den Berg
- Division of Hematology, University Hospital Basel, Basel, Switzerland
- Innovation Focus Cell Therapies, University Hospital Basel, Basel, Switzerland
| | - Heinz Läubli
- Innovation Focus Cell Therapies, University Hospital Basel, Basel, Switzerland
- Department of Biomedicine, University of Basel, Basel, Switzerland
- Division of Medical Oncology, University Hospital Basel, Basel, Switzerland
| | - Nina Khanna
- Innovation Focus Cell Therapies, University Hospital Basel, Basel, Switzerland
- Department of Biomedicine, University of Basel, Basel, Switzerland
- Division of Infectious Diseases and Hospital Epidemiology, University Hospital Basel, Basel, Switzerland
| | - Lukas T Jeker
- Innovation Focus Cell Therapies, University Hospital Basel, Basel, Switzerland
- Department of Biomedicine, University of Basel, Basel, Switzerland
- Transplantation Immunology and Nephrology, University Hospital Basel, Basel, Switzerland
| | - Andreas Holbro
- Division of Hematology, University Hospital Basel, Basel, Switzerland
- Innovation Focus Cell Therapies, University Hospital Basel, Basel, Switzerland
- Regional Blood Transfusion Service, Swiss Red Cross, Basel, Switzerland
| |
Collapse
|
41
|
Park JH, Palomba ML, Perica K, Devlin SM, Shah G, Dahi PB, Lin RJ, Salles G, Scordo M, Nath K, Valtis YK, Lynch A, Cathcart E, Zhang H, Schoder H, Leithner D, Liotta K, Yu A, Stocker K, Li J, Dey A, Sellner L, Singh R, Sundaresan V, Tong X, Zhao F, Mansilla-Soto J, He C, Meyerson J, Hosszu K, McAvoy D, Wang X, Rivière I, Sadelain M. Results From First-in-Human Phase I Study of a Novel CD19-1XX Chimeric Antigen Receptor With Calibrated Signaling in Large B-Cell Lymphoma. J Clin Oncol 2025:JCO2402424. [PMID: 39883889 DOI: 10.1200/jco-24-02424] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2024] [Revised: 11/12/2024] [Accepted: 12/13/2024] [Indexed: 02/01/2025] Open
Abstract
PURPOSE We designed a CD19-targeted chimeric antigen receptor (CAR) comprising a calibrated signaling module, termed 1XX, that differs from that of conventional CD28/CD3ζ and 4-1BB/CD3ζ CARs. Preclinical data demonstrated that 1XX CARs generated potent effector function without undermining T-cell persistence. We hypothesized that 1XX CAR T cells may be effective at low doses and elicit minimal toxicities. METHODS In this first-in-human, phase I, dose escalation and expansion clinical trial, patients with relapsed or refractory large B-cell lymphoma received 19(T2)28z-1XX CAR T cells at four dose levels (DLs), ranging from 25 to 200 × 106. RESULTS Twenty-eight patients underwent apheresis and received CAR T cells. Sixteen and 12 patients were treated in the dose escalation and expansion cohorts, respectively. The overall response rate (ORR) was 82% and complete response (CR) rate was 71% in the entire cohort. The lowest dose of 25 × 106 was selected for dose expansion. In 16 patients treated at this DL, 88% achieved ORR and 75% CR. With the median follow-up of 24 months, the 1-year event-free survival was 61% (95% CI, 45 to 82) and 14 patients remain in continuous CR beyond 12 months. In all cohorts, grade ≥3 cytokine release syndrome and immune effector cell-associated neurotoxicity syndrome rates were low at 4% and 7%, respectively. 1XX CAR T-cell products contain a higher proportion of CD8 T cells with memory features, and CAR T-cell persistence has been detected beyond 1-2 years in patients with ongoing remission. CONCLUSION The calibrated potency of the 1XX CAR affords excellent efficacy at low cell doses with favorable toxicity profiles and may benefit the treatment of other hematologic malignancies, solid tumors, and autoimmunity.
Collapse
Affiliation(s)
- Jae H Park
- Cellular Therapy Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY
- Department of Medicine, Weill Cornell Medicine, New York, NY
- Center for Cell Engineering, Sloan Kettering Institute, New York, NY
| | - M Lia Palomba
- Cellular Therapy Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY
- Department of Medicine, Weill Cornell Medicine, New York, NY
- Lymphoma Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY
| | - Karlo Perica
- Cellular Therapy Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY
- Department of Medicine, Weill Cornell Medicine, New York, NY
- Center for Cell Engineering, Sloan Kettering Institute, New York, NY
| | - Sean M Devlin
- Department of Epidemiology & Biostatistics, Memorial Sloan Kettering Cancer Center, New York, NY
| | - Gunjan Shah
- Cellular Therapy Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY
- Department of Medicine, Weill Cornell Medicine, New York, NY
- Adult Bone Marrow Transplantation Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY
| | - Parastoo B Dahi
- Cellular Therapy Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY
- Department of Medicine, Weill Cornell Medicine, New York, NY
- Adult Bone Marrow Transplantation Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY
| | - Richard J Lin
- Cellular Therapy Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY
- Department of Medicine, Weill Cornell Medicine, New York, NY
- Adult Bone Marrow Transplantation Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY
| | - Gilles Salles
- Cellular Therapy Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY
- Department of Medicine, Weill Cornell Medicine, New York, NY
- Lymphoma Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY
| | - Michael Scordo
- Cellular Therapy Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY
- Department of Medicine, Weill Cornell Medicine, New York, NY
- Adult Bone Marrow Transplantation Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY
| | - Karthik Nath
- Cellular Therapy Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY
| | - Yannis K Valtis
- Cellular Therapy Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY
| | - Alec Lynch
- Cellular Therapy Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY
| | - Elizabeth Cathcart
- Cellular Therapy Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY
| | - Honglei Zhang
- Molecular Imaging and Therapy Service, Department of Radiology, Memorial Sloan Kettering Cancer Center, New York, NY
| | - Heiko Schoder
- Molecular Imaging and Therapy Service, Department of Radiology, Memorial Sloan Kettering Cancer Center, New York, NY
| | - Doris Leithner
- Molecular Imaging and Therapy Service, Department of Radiology, Memorial Sloan Kettering Cancer Center, New York, NY
| | - Kelly Liotta
- Cellular Therapy Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY
| | - Alina Yu
- Cellular Therapy Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY
| | - Kelsey Stocker
- Cellular Therapy Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY
| | - Jia Li
- Takeda Development Center Americas, Inc, Cambridge, MA
| | - Agnish Dey
- Takeda Development Center Americas, Inc, Cambridge, MA
| | | | - Reshma Singh
- Takeda Development Center Americas, Inc, Cambridge, MA
| | | | - Xin Tong
- Takeda Development Center Americas, Inc, Cambridge, MA
| | - Faye Zhao
- Takeda Development Center Americas, Inc, Cambridge, MA
| | | | | | - Joel Meyerson
- Takeda Development Center Americas, Inc, Cambridge, MA
| | - Kinga Hosszu
- Department of Pediatrics and Immune Discovery and Modeling Service, Memorial Sloan Kettering Cancer Center, New York, NY
| | - Devin McAvoy
- Department of Pediatrics and Immune Discovery and Modeling Service, Memorial Sloan Kettering Cancer Center, New York, NY
| | - Xiuyan Wang
- Center for Cell Engineering, Sloan Kettering Institute, New York, NY
- Michael G. Harris Cell Therapy and Cell Engineering Facility, Memorial Sloan Kettering Cancer Center, New York, NY
| | - Isabelle Rivière
- Center for Cell Engineering, Sloan Kettering Institute, New York, NY
- Michael G. Harris Cell Therapy and Cell Engineering Facility, Memorial Sloan Kettering Cancer Center, New York, NY
| | - Michel Sadelain
- Center for Cell Engineering, Sloan Kettering Institute, New York, NY
| |
Collapse
|
42
|
Walder J, Soto-Lanza F, Feng L, Ahmed S, Neelapu S, Ayers A, Grosu HB, Faiz SA, Nastoupil L, Sheshadri A. Role of Pulmonary Function Tests to Predict Complications After Chimeric Antigen Receptor T-Cell Therapy. CLINICAL LYMPHOMA, MYELOMA & LEUKEMIA 2025:S2152-2650(25)00037-0. [PMID: 40000355 DOI: 10.1016/j.clml.2025.01.019] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/04/2024] [Revised: 01/18/2025] [Accepted: 01/26/2025] [Indexed: 02/27/2025]
Abstract
BACKGROUND Chimeric antigen receptor T-cell therapy (CAR-T) has transformed the treatment of certain hematologic malignancies, but toxicities limit efficacy. The role of pre-CAR-T pulmonary function testing (PFT) to predict toxicities is unclear. OBJECTIVE Our aim was to examine the association between PFTs obtained prior to CAR-T and subsequent complications in patients with lymphoma. STUDY DESIGN We conducted a retrospective study of patients who underwent standard-of-care CAR-T at our institution with pretherapy PFTs. Race-neutral normative equations from the Global Lung Initiative were used to generate percent-predicted values (PPV) for spirometry and diffusing capacity of the lungs for carbon monoxide (DLCO). Lung function score (LFS) was calculated by combining the forced expiratory volume in the first second (FEV1) and the diffusion capacity of the lung for carbon monoxide corrected for hemoglobin level (cDLCO) in an equally distributed manner, with higher score denoting worse lung function. Binary logistic regression models were used to compare the association of PFTs with complications after CAR-T. Cox regression models were additionally fit to identify predictors of mortality. RESULTS Of 218 individuals who underwent CAR-T therapy, 66 had PFTs performed within 12 months prior to lymphodepletion. Higher LFS was associated with higher risk of CRS (OR 4.3, 95% CI, 1.4-29, P = .048), after adjusting for lines of treatment. When adjusting for lines of treatment, both FEV1 (OR = 0.96, 95% CI, 0.93-0.99, P = .05) and FVC (OR = 0.96, 95% CI 0.92-0.99, P = .03) are protective for ICANS. PFT abnormalities were not associated with early or late mortality. CONCLUSION The combination of pre-CAR-T spirometry and cDLCO may help clinicians understand the risk for toxicities after CAR-T cell therapy.
Collapse
Affiliation(s)
- Jeremy Walder
- Divisions of Pulmonary, Critical Care and Sleep Medicine, McGovern Medical School at The University of Texas Health Science Center, Houston, TX
| | - Felipe Soto-Lanza
- Department of Pulmonary Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX
| | - Lei Feng
- Department of Biostatistics, The University of Texas MD Anderson Cancer Center, Houston, TX
| | - Sairah Ahmed
- Department of Stem Cell Transplantation, The University of Texas MD Anderson Cancer Center, Houston, TX; Department of Lymphoma Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX
| | - Sattva Neelapu
- Department of Lymphoma Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX
| | - Amy Ayers
- Department of Lymphoma Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX
| | - Horiana B Grosu
- Department of Pulmonary Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX
| | - Saadia A Faiz
- Department of Pulmonary Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX
| | - Loretta Nastoupil
- Department of Lymphoma Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX
| | - Ajay Sheshadri
- Department of Pulmonary Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX.
| |
Collapse
|
43
|
Ghilardi G, Hasanali ZS, Susanibar-Adaniya SP, Winestone LE, Ruella M, Garfall AL. Association of age, race, and ethnicity with access, response, and toxicities from CAR-T therapy in children and adults with B-cell malignancies: a review. J Immunother Cancer 2025; 13:e009349. [PMID: 39855710 PMCID: PMC11883890 DOI: 10.1136/jitc-2024-009349] [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: 04/10/2024] [Accepted: 01/06/2025] [Indexed: 01/27/2025] Open
Abstract
Chimeric antigen receptor T cell (CAR-T) therapies are now standard-of-care for several B-cell malignancies, and additional indications are being evaluated. In this review, we survey data on how outcomes after CAR-T therapies vary according to age, race, and ethnicity. We also review the representation of age, racial, and ethnic groups in key CAR-T clinical trials. We focus on B-cell acute lymphoblastic leukemia, B-cell non-Hodgkin's lymphoma, and multiple myeloma.
Collapse
Affiliation(s)
- Guido Ghilardi
- Center for Cellular Immunotherapies, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Zainul S Hasanali
- Department of Medicine and Abramson Cancer Center, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Sandra P Susanibar-Adaniya
- Department of Medicine and Abramson Cancer Center, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Lena E Winestone
- Division of Allergy, Immunology, and BMT, Department of Pediatrics, University of California San Francisco, San Francisco, California, USA
| | - Marco Ruella
- Center for Cellular Immunotherapies, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
- Department of Medicine and Abramson Cancer Center, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Alfred L Garfall
- Department of Medicine and Abramson Cancer Center, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| |
Collapse
|
44
|
Lepik KV, Markelov VV. The Role of the Tumor Microenvironment in T-Cell Redirecting Therapies of Large B-Cell Lymphoma: Lessons Learned from CAR-T to Bispecific Antibodies. Cancers (Basel) 2025; 17:317. [PMID: 39858099 PMCID: PMC11763497 DOI: 10.3390/cancers17020317] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2024] [Revised: 01/08/2025] [Accepted: 01/14/2025] [Indexed: 01/27/2025] Open
Abstract
T-cell redirecting therapies, which include chimeric antigen receptor T-cells (CAR-Ts) and bispecific antibodies (BSAs), have revolutionized the treatment of relapsed\refractory large B-cell lymphoma (LBCL). Expanding clinical experience with these advanced therapies shows the potential for the optimization of their use with combination or consolidation strategies, which necessitates the prognostic stratification of patients. While traditional clinical prognostic factors identified in the era of chemotherapy are characterized by limited value, the tumor microenvironment (TME) is becoming a new prognostic cluster. We examine how the heterogeneity of LBCL, characterized by variations in tumor parameters and differences in TME immune cell composition, immune checkpoint expression, and cytokine milieu, correlates with both positive responses and resistance to treatment. While classical parameters such as histological subtype, cell of origin, and target antigen expression lack proven prognostic value for T-cell redirecting therapies, the density and functional state of tumor-infiltrating lymphocytes, tumor-associated macrophages, and immune checkpoint molecules are shown to be critical determinants of therapeutic success, particularly in CAR-T therapy. We identify several gaps in the current knowledge and suggest that the insights gained from CAR-T experience could be instrumental in refining BSA applications. This report also highlights limitations in the current knowledge, as TME data derive from a limited number of registrational trials with varying methodologies, complicating cross-study comparisons and often focusing on immediate response metrics rather than long-term outcomes. By dissecting the complex interactions within the TME, this review aims to identify new prognostic factors and targets, ultimately fostering more effective and tailored treatment strategies for LBCL patients.
Collapse
Affiliation(s)
- Kirill V. Lepik
- RM Gorbacheva Research Institute of Pediatric Oncology, Hematology and Transplantation, Pavlov University, 191144 St. Petersburg, Russia
| | | |
Collapse
|
45
|
Wallace DS, Loh KP, Casulo C. How I treat older patients with relapsed/refractory diffuse large B-cell lymphoma. Blood 2025; 145:277-289. [PMID: 39356892 DOI: 10.1182/blood.2024024788] [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: 06/28/2024] [Revised: 08/19/2024] [Accepted: 09/08/2024] [Indexed: 10/04/2024] Open
Abstract
ABSTRACT Diffuse large B-cell lymphoma (DLBCL) is an aggressive, yet curable, malignancy, but older patients are at higher risk of relapsed disease because they may not be eligible for full-intensity frontline chemoimmunotherapy or have comorbidities that limit standard treatments. Recent years have brought more treatment options than ever for this patient population, but it remains challenging to determine which can be safely and effectively offered to older patients. Formal determinations of fitness including geriatric assessments remain critical, but there is less guidance on how to best use this tool in the relapsed setting. Chimeric antigen receptor T-cell therapy is accessible to older patients, provided they can be supported through the intensive road to this treatment. If relapse occurs despite this or alternative therapies are preferred, many novel therapeutic options and combinations exist with some potential modifications for older adults, such as bispecific antibodies, tafasitamab and lenalidomide, polatuzumab-containing regimens, or loncastuximab tesirine. This article provides a summary of our approach to the management of this diverse population of older patients with relapsed or refractory DLBCL.
Collapse
Affiliation(s)
- Danielle S Wallace
- Division of Hematology/Oncology, Department of Medicine, Wilmot Cancer Institute, University of Rochester Medical Center, Rochester, NY
| | - Kah Poh Loh
- Division of Hematology/Oncology, Department of Medicine, Wilmot Cancer Institute, University of Rochester Medical Center, Rochester, NY
| | - Carla Casulo
- Division of Hematology/Oncology, Department of Medicine, Wilmot Cancer Institute, University of Rochester Medical Center, Rochester, NY
| |
Collapse
|
46
|
Isshiki Y, Chen X, Teater M, Karagiannidis I, Nam H, Cai W, Meydan C, Xia M, Shen H, Gutierrez J, Easwar Kumar V, Carrasco SE, Ouseph MM, Yamshon S, Martin P, Griess O, Shema E, Porazzi P, Ruella M, Brentjens RJ, Inghirami G, Zappasodi R, Chadburn A, Melnick AM, Béguelin W. EZH2 inhibition enhances T cell immunotherapies by inducing lymphoma immunogenicity and improving T cell function. Cancer Cell 2025; 43:49-68.e9. [PMID: 39642889 PMCID: PMC11732734 DOI: 10.1016/j.ccell.2024.11.006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/21/2024] [Revised: 10/02/2024] [Accepted: 11/13/2024] [Indexed: 12/09/2024]
Abstract
T cell-based immunotherapies have demonstrated effectiveness in treating diffuse large B cell lymphoma (DLBCL) and follicular lymphoma (FL) but predicting response and understanding resistance remains a challenge. To address this, we developed syngeneic models reflecting the genetics, epigenetics, and immunology of human FL and DLBCL. We show that EZH2 inhibitors reprogram these models to re-express T cell engagement genes and render them highly immunogenic. EZH2 inhibitors do not harm tumor-controlling T cells or CAR-T cells. Instead, they reduce regulatory T cells, promote memory chimeric antigen receptor (CAR) CD8 phenotypes, and reduce exhaustion, resulting in a decreased tumor burden. Intravital 2-photon imaging shows increased CAR-T recruitment and interaction within the tumor microenvironment, improving lymphoma cell killing. Therefore, EZH2 inhibition enhances CAR-T cell efficacy through direct effects on CAR-T cells, in addition to rendering lymphoma B cells immunogenic. This approach is currently being evaluated in two clinical trials, NCT05934838 and NCT05994235, to improve immunotherapy outcomes in B cell lymphoma patients.
Collapse
MESH Headings
- Enhancer of Zeste Homolog 2 Protein/antagonists & inhibitors
- Humans
- Animals
- Mice
- Lymphoma, Large B-Cell, Diffuse/immunology
- Lymphoma, Large B-Cell, Diffuse/therapy
- Lymphoma, Large B-Cell, Diffuse/pathology
- Tumor Microenvironment/immunology
- Tumor Microenvironment/drug effects
- Lymphoma, Follicular/immunology
- Lymphoma, Follicular/therapy
- Lymphoma, Follicular/pathology
- T-Lymphocytes/immunology
- T-Lymphocytes/drug effects
- Immunotherapy, Adoptive/methods
- Immunotherapy/methods
- Cell Line, Tumor
- Receptors, Chimeric Antigen/immunology
Collapse
Affiliation(s)
- Yusuke Isshiki
- Division of Hematology/Oncology, Department of Medicine, Weill Cornell Medicine, Cornell University, New York, NY, USA
| | - Xi Chen
- Division of Hematology/Oncology, Department of Medicine, Weill Cornell Medicine, Cornell University, New York, NY, USA
| | - Matt Teater
- Division of Hematology/Oncology, Department of Medicine, Weill Cornell Medicine, Cornell University, New York, NY, USA; Institute for Computational Biomedicine, Weill Cornell Medicine, Cornell University, New York, NY, USA
| | - Ioannis Karagiannidis
- Division of Hematology/Oncology, Department of Medicine, Weill Cornell Medicine, Cornell University, New York, NY, USA
| | - Henna Nam
- Division of Hematology/Oncology, Department of Medicine, Weill Cornell Medicine, Cornell University, New York, NY, USA
| | - Winson Cai
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Cem Meydan
- Institute for Computational Biomedicine, Weill Cornell Medicine, Cornell University, New York, NY, USA; Department of Physiology and Biophysics, Weill Cornell Medicine, Cornell University, New York, NY, USA
| | - Min Xia
- Division of Hematology/Oncology, Department of Medicine, Weill Cornell Medicine, Cornell University, New York, NY, USA
| | - Hao Shen
- Division of Hematology/Oncology, Department of Medicine, Weill Cornell Medicine, Cornell University, New York, NY, USA
| | - Johana Gutierrez
- Division of Hematology/Oncology, Department of Medicine, Weill Cornell Medicine, Cornell University, New York, NY, USA
| | - Vigneshwari Easwar Kumar
- Division of Hematology/Oncology, Department of Medicine, Weill Cornell Medicine, Cornell University, New York, NY, USA
| | - Sebastián E Carrasco
- Department of Pathology and Laboratory Medicine, Weill Cornell Medicine, New York City, NY, USA; Laboratory of Comparative Pathology, Memorial Sloan Kettering Cancer Center, Weill Cornell Medicine, and Rockefeller University, New York City, NY, USA
| | - Madhu M Ouseph
- Department of Pathology and Laboratory Medicine, Weill Cornell Medicine, New York City, NY, USA
| | - Samuel Yamshon
- Division of Hematology/Oncology, Department of Medicine, Weill Cornell Medicine, Cornell University, New York, NY, USA
| | - Peter Martin
- Division of Hematology/Oncology, Department of Medicine, Weill Cornell Medicine, Cornell University, New York, NY, USA
| | - Ofir Griess
- Department of Immunology and Regenerative Biology, Weizmann Institute of Science, Rehovot 76100, Israel
| | - Efrat Shema
- Department of Immunology and Regenerative Biology, Weizmann Institute of Science, Rehovot 76100, Israel
| | - Patrizia Porazzi
- Division of Hematology and Oncology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Marco Ruella
- Division of Hematology and Oncology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Renier J Brentjens
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA; Roswell Park Comprehensive Cancer Center, Buffalo, NY, USA
| | - Giorgio Inghirami
- Department of Pathology and Laboratory Medicine, Weill Cornell Medicine, New York City, NY, USA
| | - Roberta Zappasodi
- Division of Hematology/Oncology, Department of Medicine, Weill Cornell Medicine, Cornell University, New York, NY, USA
| | - Amy Chadburn
- Department of Pathology and Laboratory Medicine, Weill Cornell Medicine, New York City, NY, USA
| | - Ari M Melnick
- Division of Hematology/Oncology, Department of Medicine, Weill Cornell Medicine, Cornell University, New York, NY, USA
| | - Wendy Béguelin
- Division of Hematology/Oncology, Department of Medicine, Weill Cornell Medicine, Cornell University, New York, NY, USA.
| |
Collapse
|
47
|
Navab R, Futela P, Kumari V, Valecha J, Gupta RB, Jain R. Advancing Multiple Myeloma Immunotherapy: A Review of Chimeric Antigen Receptor T-Cell and Bispecific T-Cell Engagers Cell Therapies in Revolutionizing Treatment. IRANIAN JOURNAL OF MEDICAL SCIENCES 2025; 50:1-10. [PMID: 39957814 PMCID: PMC11829063 DOI: 10.30476/ijms.2024.101739.3446] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/25/2024] [Revised: 05/16/2024] [Accepted: 07/07/2024] [Indexed: 02/18/2025]
Abstract
Multiple Myeloma (MM) is a hematologic malignancy characterized by clonal plasma cell development, leading to serious complications. Despite traditional treatments, MM remains incurable, necessitating innovative therapeutic approaches. Chimeric Antigen Receptor (CAR) T-cell therapy and Bispecific T-cell engagers (BiTEs) are emerging immunotherapies showing promise in MM treatment. CAR T-cell therapy involves modifying patient T-cells to target specific antigens, primarily B Cell Maturation Antigen (BCMA). BiTEs, on the other hand, are non-IgG-like bispecific antibodies designed to engage both CD3 and tumor-associated antigens. These therapies exhibit impressive efficacy in clinical trials, leading to FDA approvals for specific MM patient populations. Despite their successes, these therapies come with unique challenges and adverse effects, such as cytokine release syndrome (CRS) and neurotoxicity. This narrative review explores the mechanisms, efficacy, challenges, and potential benefits of CAR T-cell and BiTE therapies for MM patients, shedding light on their roles in addressing this complex disease.
Collapse
Affiliation(s)
- Rahul Navab
- PES Institute of Medical Sciences and Research, Kuppam, India
| | | | - Verkha Kumari
- Liaquat National Hospital and Medical College, Karachi, Pakistan
| | | | | | - Rohit Jain
- Penn State Health Milton S. Hershey Medical Center, Pennsylvania, United States of America
| |
Collapse
|
48
|
Perez-Lamas L, Sandoval-Sus J, Chavez JC. Should CAR-T cell therapy be considered a standard of care for patients with refractory diffuse large B-cell lymphoma in second line treatment? Expert Opin Biol Ther 2025; 25:139-148. [PMID: 39784146 DOI: 10.1080/14712598.2025.2451888] [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: 08/14/2024] [Revised: 11/28/2024] [Accepted: 01/07/2025] [Indexed: 01/12/2025]
Abstract
INTRODUCTION CAR-T therapy has transformed the treatment landscape for relapsed/refractory diffuse large B-cell lymphomas (DLBCL). AREAS COVERED This article reviews the existing evidence for using CAR-T therapy as a second-line treatment. Two major phase 3 trials, ZUMA-7 and TRANSFORM, have shown that axi-cel and liso-cel, respectively, offer superior outcomes compared to historical standard chemoimmunotherapy and consolidation with autologous hematopoietic stem cell transplantation (auto-HCT). Additionally, two promising phase 2 trials, PILOT and ALYCANTE, demonstrated the efficacy of CAR-T therapy in patients who are ineligible for auto-HCT. We also reviewed the potential biological factors behind these results. EXPERT OPINION Several factors support the use of CAR-T therapy in earlier treatment lines: better T-cell fitness in the infused product, reduced systemic inflammation in patients, and a more favorable tumor microenvironment. Although real-world data for second-line CAR-T therapy is still early, it is expected that CAR-T will be used more widely. Additional focus highlights the need for defining suitable patient populations and the efforts to enhance accessibility and cost-effectiveness of this groundbreaking treatment approach.
Collapse
Affiliation(s)
| | - Jose Sandoval-Sus
- Malignant Hematology and Cellular Therapy, Memorial Health Care, Pembroke Pines, USA
| | - Julio C Chavez
- Department of Malignant Hematology, Moffitt Cancer Center, Tampa, FL, USA
| |
Collapse
|
49
|
Bisio M, Legato L, Fasano F, Benevolo Savelli C, Boccomini C, Nicolosi M, Santambrogio E, Freilone R, Novo M, Botto B. Bispecific Antibodies for Lymphoid Malignancy Treatment. Cancers (Basel) 2024; 17:94. [PMID: 39796723 PMCID: PMC11719988 DOI: 10.3390/cancers17010094] [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: 10/14/2024] [Revised: 11/30/2024] [Accepted: 12/10/2024] [Indexed: 01/13/2025] Open
Abstract
BACKGROUD The introduction of highly active immunotherapies has changed the outcome of B-cell non-Hodgkin lymphomas (B-NHLs) in the last two decades. Since then, important progress has been shown using newer and more active immunotherapies, including chimeric antigen receptor T-cell therapy (CAR-T), conjugated monoclonal antibodies, and bispecific antobodies, which currently plays a significant role in the treatment of diffuse large B-cell (DLBCL), follicular (FL), and mantle cell (MCL) lymphoma. PURPOSE In this review, we provide an updated overview of recently completed and ongoing BsAb trials in patients with relapsed/refractory(R/R) B-NHL and Hodgkin's lymphoma, including single-agent results, emerging combinations, safety data, and novel constructs. CONCLUSIONS Bispecific antibodies (BsAbs) are a novel class of "off-the-shelf" T-cell-redirecting drugs capable of targeting various cell-surface antigens. New antigen targets are currently under investigation, such as CD19 × CD3 and CD30 × CD3 or CD30 × CD16, in different settings. BsAbs are among the most promising therapeutic options for lymphoma today since they have demonstrated significant single-agent activity, along with a manageable toxicity profile, in patients with heavily pretreated B-NHL.
Collapse
Affiliation(s)
- Matteo Bisio
- Hematology Division, A.O.U. Città della Salute e della Scienza di Torino, C.so Bramante 88, 10126 Turin, Italy (R.F.); (M.N.)
| | | | | | | | | | | | | | | | | | - Barbara Botto
- Hematology Division, A.O.U. Città della Salute e della Scienza di Torino, C.so Bramante 88, 10126 Turin, Italy (R.F.); (M.N.)
| |
Collapse
|
50
|
Yang J, Song Y, Zhou K, Li Z, Zhang M, Jing H, Wang Z, Yu L, Meng W, Lu Q, Tian W, Shi Y. Safety and efficacy of amulirafusp alfa (IMM0306), a fusion protein of CD20 monoclonal antibody with the CD47 binding domain of SIRPα, in patients with relapsed or refractory B-cell non-Hodgkin lymphoma: a phase 1/2 study. J Hematol Oncol 2024; 17:123. [PMID: 39696680 PMCID: PMC11657391 DOI: 10.1186/s13045-024-01646-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2024] [Accepted: 11/28/2024] [Indexed: 12/20/2024] Open
Abstract
BACKGROUND Amulirafusp alfa (IMM0306) is a fusion protein of CD47 binding domain of signal-regulatory protein alpha (SIRPα) with CD20 monoclonal antibody on both heavy chains. This study aimed to evaluate the safety and preliminary efficacy of amulirafusp alfa in relapsed or refractory (r/r) B-cell non-Hodgkin lymphoma (B-NHL). METHODS We enrolled patients with CD20 + r/r B-NHL who had previously received at least two lines of therapy to receive a single-dose of amulirafusp alfa in the first 2 weeks, followed by a multiple-dose period, in which the patients received the same intravenous dose every week in 4-week cycles. The primary endpoints were to evaluate the safety, determine the maximum tolerated dose (MTD) and the recommended phase 2 dose (RP2D) of amulirafusp alfa. RESULTS Between May 22, 2020 and February 10, 2022, 48 patients with r/r B-NHL were enrolled and received amulirafusp alfa at the doses of 40-2000 μg/kg. As of the data cut-off date of April 18, 2024, no dose-limiting toxicity was observed, and the MTD was not reached. The dose of 2000 μg/kg was identified as the RP2D. All grades and ≥ grade 3 treatment-related adverse events (TRAEs) occurred in 48 (100%) and 33 (68.8%) patients, respectively. The most common ≥ grade 3 TRAEs were lymphocyte count decreased (28/48, 58.3%), white blood cell count decreased (10/48, 20.8%), absolute neutrophil count decreased (9/48, 18.8%) and anemia (5/48, 10.4%). At the doses of 800-2000 μg/kg, objective response rate in follicular lymphoma and marginal zone lymphoma was 41.2% (7/17, 95% confidence interval [CI] 18.4-67.1) and 33.3% (2/6, 95% CI 3.7-71.0), respectively. CONCLUSION Amulirafusp alfa showed favorable safety profile and preliminary efficacy in patients with r/r B-NHL, meriting further investigation. Trial registration NCT05805943.
Collapse
Affiliation(s)
- Jianliang Yang
- Department of Medical Oncology, Beijing Key Laboratory of Clinical Study on Anticancer Molecular Targeted Drugs, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, No. 17 Panjiayuan Nanli, Chaoyang District, Beijing, 100021, China
| | - Yongping Song
- Department of Hematology, Affiliated Cancer Hospital of Zhengzhou University, Henan Cancer Hospital, Zhengzhou, China
| | - Keshu Zhou
- Department of Hematology, Affiliated Cancer Hospital of Zhengzhou University, Henan Cancer Hospital, Zhengzhou, China
| | - Zhiming Li
- Department of Medical Oncology, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-Sen University Cancer Center, Guangzhou, China
| | - Mingzhi Zhang
- Department of Oncology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Hongmei Jing
- Department of Hematology, Peking University Third Hospital, Beijing, China
| | - Zhen Wang
- Department of Medical Oncology, Linyi Cancer Hospital, Linyi, China
| | - Li Yu
- Department of Hematology, The Second Affiliated Hospital of Nanchang University, Nanchang, China
| | - Wei Meng
- ImmuneOnco Biopharmaceuticals (Shanghai) Inc., Shanghai, China
| | - Qiying Lu
- ImmuneOnco Biopharmaceuticals (Shanghai) Inc., Shanghai, China
| | - Wenzhi Tian
- ImmuneOnco Biopharmaceuticals (Shanghai) Inc., Shanghai, China
| | - Yuankai Shi
- Department of Medical Oncology, Beijing Key Laboratory of Clinical Study on Anticancer Molecular Targeted Drugs, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, No. 17 Panjiayuan Nanli, Chaoyang District, Beijing, 100021, China.
| |
Collapse
|