1
|
Ailawadhi S, Biru Y, Clavreul S, San Miguel M, Cormier N, Efebera Y, Merz M, Sato A, Zeanah C, Watkins JL, Farrell J, Goldman EH, Popat R. Perspectives of Healthcare Providers and Patients with Relapsed/Refractory Multiple Myeloma on Treatment Priorities and Novel Therapies. Patient Prefer Adherence 2025; 19:1089-1104. [PMID: 40260187 PMCID: PMC12010075 DOI: 10.2147/ppa.s496106] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/21/2024] [Accepted: 04/02/2025] [Indexed: 04/23/2025] Open
Abstract
Purpose With novel therapies including chimeric antigen receptor T-cell (CAR-T) therapy and bispecific antibodies (BsAbs), healthcare providers (HCPs) face complexities managing treatment for patients with relapsed/refractory multiple myeloma (RRMM). This study, among the largest surveys on RRMM, examined unmet needs in care access, barriers to novel therapy use, and treatment decision-making. Methods This survey-based study (March-June 2024) enrolled 2284 participants (patients: 1301; HCPs: 983) across 7 countries. Patients with >1 relapse/progression and HCPs managing ≥3 patients were included. Data were analyzed using descriptive statistics and Χ2 tests. Results For patients, treatment priorities included slowing disease progression (second line [2L], 47%; third or later line [≥3L], 49%), minimizing adverse events (AEs; 2L, 43%; ≥3L, 49%), and extending life (2L, 39%; ≥3L, 38%). HCPs prioritized prolonging survival and controlling disease. Younger patients (<65 vs ≥65 years) prioritized convenience (40% vs 24%; P<0.01) and avoiding referrals to new institutions for therapies (32% vs 20%; P<0.01). Across geographies, HCPs reported logistical challenges as key reasons that CAR-T (38%) or BsAb (34%) therapy was not offered. Novel therapies were offered to patients more frequently in the US vs EU (CAR-T, 84% vs 77%, P=0.023; BsAbs, 84% vs 76%, P=0.011), with a similar trend in the US vs Japan for CAR-T; however, across all geographies, few patients recalled being offered CAR-T (17%) or BsAbs (13%). Patients receiving BsAbs prioritized efficacy-related reasons (25-35%) and nonclinical factors like less time and financial impact (27-29%), whereas those who received CAR-T prioritized patient success stories (50%), efficacy-related factors (48-50%), and minimal financial burden (43%). Conclusion This study revealed gaps in treatment priorities; patients valued quality of life and AE management, while HCPs focused on efficacy and delaying progression. There is a significant need to educate HCPs and patients on the impact of shared decision-making when considering novel treatments for RRMM.
Collapse
Affiliation(s)
- Sikander Ailawadhi
- Division of Hematology and Medical Oncology, Mayo Clinic, Jacksonville, FL, USA
| | - Yelak Biru
- International Myeloma Foundation, Studio City, CA, USA
| | | | | | | | | | - Maximilian Merz
- Multiple Myeloma Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA; Department of Hematology and Cell Therapy, University of Leipzig, Leipzig, Germany
| | | | | | | | | | | | - Rakesh Popat
- National Institute for Health Research Clinical Research Facility, University College London Hospitals NHS Trust, London, UK
| |
Collapse
|
2
|
Shahid S, Prockop SE, Flynn GC, Mauguen A, White CO, Bieler J, McAvoy D, Hosszu K, Cancio MI, Jakubowski AA, Scaradavou A, Boelens JJ, Sauter CS, Perales MA, Giralt SA, Taylor C, Chaudhari J, Wang X, Rivière I, Sadelain M, Brentjens RJ, Kernan NA, O'Reilly RJ, Curran KJ. Allogeneic off-the-shelf CAR T-cell therapy for relapsed or refractory B-cell malignancies. Blood Adv 2025; 9:1644-1657. [PMID: 39908482 PMCID: PMC11995077 DOI: 10.1182/bloodadvances.2024015157] [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: 10/23/2024] [Revised: 12/10/2024] [Accepted: 12/31/2024] [Indexed: 02/07/2025] Open
Abstract
ABSTRACT Despite clinical benefit with the use of chimeric antigen receptor (CAR) T cells, the need to manufacture patient-specific products limits its clinical utility. To overcome this barrier, we developed an allogeneic "off-the-shelf" CAR T-cell product using Epstein-Barr virus (EBV)-specific T cells (EBV-VSTs) genetically modified with a CD19-specific CAR (19-28z). Patients with relapsed/refractory (R/R) B-cell malignancies were stratified into 3 treatment cohorts: cohort 1 (n = 8; disease recurrence after allogeneic or autologous hematopoietic cell transplantation [HCT]), cohort 2 (n = 6; consolidative therapy after autologous HCT), or cohort 3 (n = 2; consolidative therapy after allogeneic HCT). The primary objective of this trial was to determine the safety of multiple CAR EBV-VST infusions. Most patients (n = 12/16) received multiple doses (overall median, 2.5 [range, 1-3]) with 3 × 106 T cells per kg determined to be the optimal dose enabling multiple treatments per manufactured cell line. Severe cytokine release syndrome or neurotoxicity did not occur after infusion, and no dose-limiting toxicity was observed in the trial. Median follow-up was 48 months (range, 4-135) with 4 deaths due to disease progression. Overall survival of all patients was 81% at 12 months and 75% at 36 months. Postinfusion expansion and persistence were limited, and CAR EBV-VSTs demonstrated a unique T-cell phenotype compared with autologous 19-28z CAR T cells. Our study demonstrates the feasibility and safety of an allogeneic "off-the-shelf" CAR EBV-VST product with favorable outcomes for patients with CD19+ R/R B-cell malignancies. This trial was registered at www.ClinicalTrials.gov as #NCT01430390.
Collapse
MESH Headings
- Humans
- Immunotherapy, Adoptive/methods
- Immunotherapy, Adoptive/adverse effects
- Middle Aged
- Adult
- Male
- Female
- Receptors, Chimeric Antigen/therapeutic use
- Aged
- Antigens, CD19/immunology
- T-Lymphocytes/immunology
- T-Lymphocytes/transplantation
- Transplantation, Homologous
- Hematopoietic Stem Cell Transplantation/methods
- Hematopoietic Stem Cell Transplantation/adverse effects
- Herpesvirus 4, Human
- Leukemia, B-Cell/therapy
- Treatment Outcome
- Lymphoma, B-Cell/therapy
- Lymphoma, B-Cell/immunology
- Receptors, Antigen, T-Cell/therapeutic use
- Receptors, Antigen, T-Cell/genetics
- Young Adult
Collapse
Affiliation(s)
- Sanam Shahid
- Department of Pediatrics, Memorial Sloan Kettering Cancer Center, New York, NY
| | - Susan E. Prockop
- Department of Hematopoietic Stem Cell Transplant, Dana-Farber/Boston Children's Cancer and Blood Disorders Center, Boston, MA
| | - Georgia C. Flynn
- Department of Pediatrics, Memorial Sloan Kettering Cancer Center, New York, NY
| | - Audrey Mauguen
- Department of Epidemiology-Biostatistics, Memorial Sloan Kettering Cancer Center, New York, NY
| | - Charlie O. White
- Department of Epidemiology-Biostatistics, Memorial Sloan Kettering Cancer Center, New York, NY
| | - Jennifer Bieler
- Department of Pediatrics, Memorial Sloan Kettering Cancer Center, New York, NY
| | - Devin McAvoy
- Department of Pediatrics, Memorial Sloan Kettering Cancer Center, New York, NY
| | - Kinga Hosszu
- Department of Pediatrics, Memorial Sloan Kettering Cancer Center, New York, NY
| | - Maria I. Cancio
- Department of Pediatrics, Memorial Sloan Kettering Cancer Center, New York, NY
| | - Ann A. Jakubowski
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY
| | | | - Jaap Jan Boelens
- Department of Pediatrics, Memorial Sloan Kettering Cancer Center, New York, NY
| | - Craig S. Sauter
- Department of Hematology and Medical Oncology, Taussig Cancer Institute, Cleveland Clinic, Cleveland, OH
| | | | - Sergio A. Giralt
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY
| | - Clare Taylor
- Department of Pharmacology, Cell Therapy and Cell Engineering Facility, Memorial Sloan Kettering Cancer Center, New York, NY
| | - Jagrutiben Chaudhari
- Department of Pharmacology, Cell Therapy and Cell Engineering Facility, Memorial Sloan Kettering Cancer Center, New York, NY
| | - Xiuyan Wang
- Department of Pharmacology, Cell Therapy and Cell Engineering Facility, Memorial Sloan Kettering Cancer Center, New York, NY
| | - Isabelle Rivière
- Department of Pharmacology, Cell Therapy and Cell Engineering Facility, Memorial Sloan Kettering Cancer Center, New York, NY
| | - Michel Sadelain
- Department of Pharmacology, Cell Therapy and Cell Engineering Facility, Memorial Sloan Kettering Cancer Center, New York, NY
| | - Renier J. Brentjens
- Department of Medicine, Roswell Park Comprehensive Cancer Center, Buffalo, NY
| | - Nancy A. Kernan
- Department of Pediatrics, Memorial Sloan Kettering Cancer Center, New York, NY
| | - Richard J. O'Reilly
- Department of Pediatrics, Memorial Sloan Kettering Cancer Center, New York, NY
| | - Kevin J. Curran
- Department of Pediatrics, Memorial Sloan Kettering Cancer Center, New York, NY
| |
Collapse
|
3
|
Mahmoudjafari Z, Li J, Bercaw E, Parise 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 2025:1-30. [PMID: 40163049 DOI: 10.1080/13696998.2025.2486839] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [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 fora 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
|
4
|
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
|
5
|
Jørgensen LV, Christensen EB, Barnkob MB, Barington T. The clinical landscape of CAR NK cells. Exp Hematol Oncol 2025; 14:46. [PMID: 40149002 PMCID: PMC11951618 DOI: 10.1186/s40164-025-00633-8] [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: 11/01/2024] [Accepted: 03/04/2025] [Indexed: 03/29/2025] Open
Abstract
Chimeric antigen receptor (CAR) NK cell therapy has emerged as a promising alternative to CAR T cell therapy, offering significant advantages in terms of safety and versatility. Here we explore the current clinical landscape of CAR NK cells, and their application in hematologic malignancies and solid cancers, as well as their potential for treating autoimmune disorders. Our analysis draws from data collected from 120 clinical trials focused on CAR NK cells, and presents insights into the demographics and characteristics of these studies. We further outline the specific targets and diseases under investigation, along with the major cell sources, genetic modifications, combination strategies, preconditioning- and dosing regimens, and manufacturing strategies being utilized. Initial results from 16 of these clinical trials demonstrate promising efficacy of CAR NK cells, particularly in B cell malignancies, where response rates are comparable to those seen with CAR T cells but with lower rates of severe adverse effects, such as cytokine release syndrome (CRS), immune effector cell-associated neurotoxicity syndrome (ICANS), and graft-versus-host disease (GvHD). However, challenges remain in solid tumor applications, where only modest efficacy has been observed to date. Our analysis reveals that research is increasingly focused on enhancing CAR NK cell persistence, broadening their therapeutic targets, and refining manufacturing processes to improve accessibility and scalability. With recent advancements in NK cell engineering and their increased clinical applications, CAR NK cells are predicted to become an integral component of next-generation immunotherapies, not only for cancer but potentially for immune-mediated diseases as well.
Collapse
Affiliation(s)
- Lasse Vedel Jørgensen
- Department of Clinical Immunology, Odense University Hospital, Odense, Denmark
- Centre for Cellular Immunotherapy of Haematological Cancer Odense (CITCO), Odense, Denmark
| | - Emil Birch Christensen
- Department of Clinical Immunology, Odense University Hospital, Odense, Denmark
- Centre for Cellular Immunotherapy of Haematological Cancer Odense (CITCO), Odense, Denmark
| | - Mike Bogetofte Barnkob
- Department of Clinical Immunology, Odense University Hospital, Odense, Denmark
- Centre for Cellular Immunotherapy of Haematological Cancer Odense (CITCO), Odense, Denmark
| | - Torben Barington
- Department of Clinical Immunology, Odense University Hospital, Odense, Denmark.
- Centre for Cellular Immunotherapy of Haematological Cancer Odense (CITCO), Odense, Denmark.
| |
Collapse
|
6
|
Goldmacher VS, Gershteyn I, Chari R, Kovtun Y. A bispecific anti-MUC16/anti-death receptor 5 antibody achieves effective and tumor-selective death receptor 5-mediated tumor regression. Sci Rep 2025; 15:9909. [PMID: 40121208 PMCID: PMC11929789 DOI: 10.1038/s41598-025-93927-0] [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: 01/21/2025] [Accepted: 03/10/2025] [Indexed: 03/25/2025] Open
Abstract
The bispecific antibody IMV-M was designed to selectively bind and cluster death receptor 5 (DR5) upon engaging the tumor antigen MUC16 through a novel mechanism-clustering multiple IMV-M molecules on a single MUC16 molecule. IMV-M demonstrated potent, MUC16-selective anti-tumor activity in vitro and in xenograft models without requiring secondary crosslinking, and a pilot non-human primate toxicity study detected no toxicity. Our findings suggest that antibody clustering effectively induces DR5 clustering, resulting in anti-tumor activity. Unlike anti-MUC16 antibody-drug conjugates (ADCs), which rely on cytotoxic payloads, this approach offers a safer and more effective therapeutic strategy. Notably, MUC16 is overexpressed in substantial subsets of ovarian, pancreatic, and lung cancers, with minimal expression in normal tissues, suggesting the broad applicability of this bispecific antibody.
Collapse
Affiliation(s)
- Victor S Goldmacher
- Research and Development, ImmuVia Inc, 245 First Street, Suite 1800, Cambridge, MA, 02142, USA.
| | - Iosif Gershteyn
- Research and Development, ImmuVia Inc, 245 First Street, Suite 1800, Cambridge, MA, 02142, USA
| | - Ravi Chari
- Research and Development, ImmuVia Inc, 245 First Street, Suite 1800, Cambridge, MA, 02142, USA
| | - Yelena Kovtun
- Research and Development, ImmuVia Inc, 245 First Street, Suite 1800, Cambridge, MA, 02142, USA
| |
Collapse
|
7
|
Bubb QR, Balood M, Seir GE, Swartzrock L, Haslett E, Ho K, Xu P, Wiltz SG, Sotillo E, Gruber TA, Richards RM, Mackall CL, Czechowicz A. Development of multivalent CAR T cells as dual immunotherapy and conditioning agents. MOLECULAR THERAPY. ONCOLOGY 2025; 33:200944. [PMID: 40034967 PMCID: PMC11872492 DOI: 10.1016/j.omton.2025.200944] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/11/2024] [Revised: 11/18/2024] [Accepted: 01/28/2025] [Indexed: 03/05/2025]
Abstract
Hematopoietic stem cell transplantation (HSCT) is the only definitive cure for pediatric acute myeloid leukemia (AML). Despite adjustments in HSCT protocols and improvements in supportive care, 30% of high-risk patients who receive HSCT as part of their therapy still experience disease relapse with high transplant-related mortality. Relapsed AML has a dismal prognosis, and novel therapies are needed. To improve upon the status quo, HSCT would more effectively eliminate relapse-initiating leukemic cells and be delivered with safer, non-genotoxic conditioning. Here, we investigate hematopoietic cytokine receptors (HCRs) and identify that KIT, MPL, and FLT3 are collectively highly expressed in virtually all pediatric AML samples studied. Further, we establish proof-of-concept of a first-in-class chimeric antigen receptor (CAR) T cell that enables simultaneous targeting of KIT, MPL, and FLT3 through a single receptor, which we term the extracellularly linked concatemeric trivalent cytokine (ELECTRIC) CAR. ELECTRIC CARs exhibit potent cytotoxicity against normal and malignant hematopoietic cells in vitro and display anti-HCR activity in a murine xenograft model. We propose that the ELECTRIC system can be the foundation to developing a non-genotoxic, anti-leukemic conditioning regimen to enable safer, more durable efficacy with minimal toxicity.
Collapse
Affiliation(s)
- Quenton Rashawn Bubb
- Stem Cell Biology and Regenerative Medicine Graduate Program, Medical Scientist Training Program, Stanford University School of Medicine, Stanford, CA 94305, USA
- Department of Pediatrics, Division of Hematology, Oncology, Stem Cell Transplantation and Regenerative Medicine, Stanford University School of Medicine, Stanford, CA 94305, USA
- Institute for Stem Cell Biology and Regenerative Medicine, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Mohammad Balood
- Department of Pediatrics, Division of Hematology, Oncology, Stem Cell Transplantation and Regenerative Medicine, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Gabe Eduardo Seir
- Department of Pediatrics, Division of Hematology, Oncology, Stem Cell Transplantation and Regenerative Medicine, Stanford University School of Medicine, Stanford, CA 94305, USA
- Department of Bioengineering, Stanford University, Stanford, CA 94305, USA
| | - Leah Swartzrock
- Department of Pediatrics, Division of Hematology, Oncology, Stem Cell Transplantation and Regenerative Medicine, Stanford University School of Medicine, Stanford, CA 94305, USA
- Institute for Stem Cell Biology and Regenerative Medicine, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Ethan Haslett
- Department of Pediatrics, Division of Hematology, Oncology, Stem Cell Transplantation and Regenerative Medicine, Stanford University School of Medicine, Stanford, CA 94305, USA
- Institute for Stem Cell Biology and Regenerative Medicine, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Katie Ho
- Department of Pediatrics, Division of Hematology, Oncology, Stem Cell Transplantation and Regenerative Medicine, Stanford University School of Medicine, Stanford, CA 94305, USA
- Institute for Stem Cell Biology and Regenerative Medicine, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Peng Xu
- Stanford Cancer Institute, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Saida G. Wiltz
- Department of Pediatrics, Division of Hematology, Oncology, Stem Cell Transplantation and Regenerative Medicine, Stanford University School of Medicine, Stanford, CA 94305, USA
- Institute for Stem Cell Biology and Regenerative Medicine, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Elena Sotillo
- Stanford Cancer Institute, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Tanja A. Gruber
- Department of Pediatrics, Division of Hematology, Oncology, Stem Cell Transplantation and Regenerative Medicine, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Rebecca M. Richards
- Department of Pediatrics, University of Wisconsin-Madison, Madison, WI 53792, USA
| | - Crystal L. Mackall
- Department of Pediatrics, Division of Hematology, Oncology, Stem Cell Transplantation and Regenerative Medicine, Stanford University School of Medicine, Stanford, CA 94305, USA
- Stanford Cancer Institute, Stanford University School of Medicine, Stanford, CA 94305, USA
- Division of Blood and Stem Cell Transplantation and Cell Therapy, Department of Medicine, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Agnieszka Czechowicz
- Department of Pediatrics, Division of Hematology, Oncology, Stem Cell Transplantation and Regenerative Medicine, Stanford University School of Medicine, Stanford, CA 94305, USA
- Institute for Stem Cell Biology and Regenerative Medicine, Stanford University School of Medicine, Stanford, CA 94305, USA
| |
Collapse
|
8
|
Tan YH, Yoon DH, Davies AJ, Buske C, Boo YL, Somasundaram N, Lim F, Ong SY, Jeyasekharan A, Izutsu K, Kim WS, Chan JY. Improving access to chimeric antigen receptor T-cells for refractory or relapsing diffuse large B cell lymphoma therapy in Asia. Discov Oncol 2025; 16:181. [PMID: 39951161 PMCID: PMC11828776 DOI: 10.1007/s12672-025-01860-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/20/2024] [Accepted: 02/03/2025] [Indexed: 02/17/2025] Open
Abstract
Chimeric antigen receptor T-cell (CAR-T)-mediated therapies have shown promising clinical benefit in patients with refractory or relapsing (R/R) diffuse large B-cell lymphoma (DLBCL). However, CAR-T treatment presents challenges such as lack of drug accessibility, financial barriers, variable physician preference or experience, and risk assessment based on patient-specific characteristics. This article thus aims to provide an overview of the CAR-T landscape for R/R DLBCL in Asia, with a focus on identifying barriers to access, from the perspective of Asian and international lymphoma experts. Presently, existing clinical data indicate that CAR-T therapy is a potentially curative strategy for R/R DLBCL in addition to stem cell transplantation, provided the patient's disease profile and treatment history have been thoroughly considered. However, longer-term follow-up data from large-scale studies are needed to confirm curative potential and define optimal sequencing of CAR-T in the context of novel emerging treatments, such as bi-specific antibodies, in the management of R/R DLBCL. Consequently, further research into CAR-T would benefit from collaboration between institutions. Furthermore, there is a wide disparity in CAR-T accessibility across regions due to complicated logistics and cost, which represent a significant barrier to patients in Asia. Hence, there is a need to increase representation and engagement across different stakeholders such as policymakers, payers, and the industry to arrive at a consensus on patient selection, establish clear guidelines, and develop strategies to lower CAR-T costs. Ultimately, data can support a multi-stakeholder approach when devising strategies to make CAR-T feasible and sustainable for patients.
Collapse
Affiliation(s)
- Ya Hwee Tan
- Division of Medical Oncology, National Cancer Centre Singapore, Singapore, Singapore
| | - Dok Hyun Yoon
- Department of Oncology, Asan Medical Center, University of Ulsan College of Medicine, Seoul, South Korea
| | - Andrew J Davies
- School of Cancer Sciences, Faculty of Medicine, University of Southampton, Southampton, UK
| | - Christian Buske
- Institute of Experimental Cancer Research, University Hospital of Ulm, Ulm, Germany
| | - Yang Liang Boo
- Department of Hematology, Hospital Sultanah Aminah, Johor Bahru, Malaysia
| | - Nagavalli Somasundaram
- Division of Medical Oncology, National Cancer Centre Singapore, Singapore, Singapore
- Duke-NUS Medical School, National Cancer Centre Singapore, Singapore, Singapore
| | - Francesca Lim
- Duke-NUS Medical School, National Cancer Centre Singapore, Singapore, Singapore
- Department of Haematology, Singapore General Hospital, Singapore, Singapore
| | - Shin Yeu Ong
- Department of Haematology, Singapore General Hospital, Singapore, Singapore
| | - Anand Jeyasekharan
- Department of Haematology-Oncology, National University Cancer Institute, Singapore, Singapore
- Cancer Science Institute of Singapore, National University of Singapore, Singapore, Singapore
| | - Koji Izutsu
- Department of Hematology, National Cancer Center Hospital, Tokyo, Japan
| | - Won Seog Kim
- Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, South Korea
| | - Jason Yongsheng Chan
- Division of Medical Oncology, National Cancer Centre Singapore, Singapore, Singapore.
- Duke-NUS Medical School, National Cancer Centre Singapore, Singapore, Singapore.
| |
Collapse
|
9
|
Goto H, Kumode T, Mishima Y, Kataoka K, Ogawa Y, Kanemura N, Shimada K, Uchida T, Kuroe Y, Kawasaki A, Sato J, Teshima T. Efficacy and safety of mosunetuzumab monotherapy for Japanese patients with relapsed/refractory follicular lymphoma: FLMOON-1. Int J Clin Oncol 2025; 30:389-396. [PMID: 39652156 PMCID: PMC11785659 DOI: 10.1007/s10147-024-02662-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2024] [Accepted: 11/11/2024] [Indexed: 02/01/2025]
Abstract
BACKGROUND In a global phase I/II study (GO29781; NCT02500407), single-agent mosunetuzumab had a manageable safety profile and induced durable complete responses in patients with relapsed/refractory (R/R) B-cell non-Hodgkin lymphoma, including in patients with R/R follicular lymphoma (FL). In this analysis, the efficacy and safety of mosunetuzumab monotherapy were evaluated in an expansion cohort, FLMOON-1, in Japanese patients with R/R FL who had received ≥ 2 prior lines of therapy in a phase I study (JO40295, jRCT2080223801). METHODS Mosunetuzumab was administered intravenously at the recommended phase II dose (with cycle 1 step-up dosing) for eight cycles or up to 17 cycles, or until disease progression or unacceptable toxicity. The pre-specified primary endpoint was Independent Review Facility (IRF)-assessed complete response rate (CRR; as best overall response). Secondary objectives included investigator (INV)-assessed CRR, INV- and IRF-assessed objective response rate (ORR), and safety. RESULTS At the data cutoff (October 13, 2023), 19 patients (median age 72 years) were evaluated. The IRF-assessed CRR and ORR were 68.4% and 78.9%, respectively; the INV-assessed CRR and ORR were 63.2% and 84.2%, respectively. Grade 3-4 adverse events (AEs) were observed in 89.5% of patients, with a low incidence of AEs leading to mosunetuzumab discontinuation (10.5%) and one fatal AE unrelated to mosunetuzumab. Cytokine release syndrome occurred in 47.4% of patients and were mostly Grade 1 in severity. CONCLUSION These findings indicate mosunetuzumab has a consistent efficacy and manageable safety profile in Japanese patients with R/R FL compared with previously reported data from the global phase I/II study.
Collapse
MESH Headings
- Humans
- Lymphoma, Follicular/drug therapy
- Male
- Middle Aged
- Aged
- Female
- Adult
- Japan
- Antibodies, Monoclonal, Humanized/therapeutic use
- Antibodies, Monoclonal, Humanized/adverse effects
- Antibodies, Monoclonal, Humanized/administration & dosage
- Neoplasm Recurrence, Local/drug therapy
- Aged, 80 and over
- Antineoplastic Agents, Immunological/therapeutic use
- Antineoplastic Agents, Immunological/adverse effects
- Antineoplastic Agents, Immunological/administration & dosage
- East Asian People
Collapse
Affiliation(s)
- Hideki Goto
- Department of Hematology, Faculty of Medicine and Graduate School of Medicine, Hokkaido University, Hokkaido, Japan.
- Division of Laboratory and Transfusion Medicine, Hokkaido University Hospital, Hokkaido, Japan.
| | - Takahiro Kumode
- Department of Hematology and Rheumatology, Faculty of Medicine, Kindai University, Osaka, Japan
| | - Yuko Mishima
- Department of Hematology Oncology, Japanese Foundation for Cancer Research, Cancer Institute Hospital, Tokyo, Japan
| | - Keisuke Kataoka
- Division of Hematology, Department of Medicine, Keio University School of Medicine, Tokyo, Japan
| | - Yoshiaki Ogawa
- Department of Hematology & Oncology, Tokai University School of Medicine, Kanagawa, Japan
| | - Nobuhiro Kanemura
- Department of Hematology and Infectious Disease, Gifu University Hospital, Gifu, Japan
| | - Kazuyuki Shimada
- Department of Hematology and Oncology, Nagoya University Graduate School of Medicine, Aichi, Japan
| | - Toshiki Uchida
- Department of Hematology and Oncology, Japanese Red Cross Aichi Medical Center Nagoya Daini Hospital, Aichi, Japan
| | | | | | - Jotaro Sato
- Chugai Pharmaceutical Co., Ltd, Tokyo, Japan
| | - Takanori Teshima
- Department of Hematology, Faculty of Medicine and Graduate School of Medicine, Hokkaido University, Hokkaido, Japan
- Division of Laboratory and Transfusion Medicine, Hokkaido University Hospital, Hokkaido, Japan
| |
Collapse
|
10
|
Basingab FS, Alshahrani OA, Alansari IH, Almarghalani NA, Alshelali NH, Alsaiary AH, Alharbi N, Zaher KA. From Pioneering Discoveries to Innovative Therapies: A Journey Through the History and Advancements of Nanoparticles in Breast Cancer Treatment. BREAST CANCER (DOVE MEDICAL PRESS) 2025; 17:27-51. [PMID: 39867813 PMCID: PMC11761866 DOI: 10.2147/bctt.s501448] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/17/2024] [Accepted: 01/03/2025] [Indexed: 01/28/2025]
Abstract
Nanoparticle technology has revolutionized breast cancer treatment by offering innovative solutions addressing the gaps in traditional treatment methods. This paper aimed to comprehensively explore the historical journey and advancements of nanoparticles in breast cancer treatment, highlighting their transformative impact on modern medicine. The discussion traces the evolution of nanoparticle-based therapies from their early conceptualization to their current applications and future potential. We initially explored the historical context of breast cancer treatment, highlighting the limitations of conventional therapies, such as surgery, radiation, and chemotherapy. The advent of nanotechnology has introduced a new era characterized by the development of various nanoparticles, including liposomes, dendrimers, and gold nanoparticles, designed to target cancer cells with remarkable precision. We further described the mechanisms of action for nanoparticles, including passive and active targeting, and reviewed significant breakthroughs and clinical trials that have validated their efficacy. Current applications of nanoparticles in breast cancer treatment have been examined, showcasing clinically approved therapies and comparing their effectiveness with traditional methods. This article also discusses the latest advancements in nanoparticle research, including drug delivery systems and combination therapy innovations, while addressing the current technical, biological, and regulatory challenges. The technical challenges include efficient and targeted delivery to tumor sites without affecting healthy tissue; biological, such as potential toxicity, immune system activation, or resistance mechanisms; economic, involving high production and scaling costs; and regulatory, requiring rigorous testing for safety, efficacy, and long-term effects to meet stringent approval standards. Finally, we have explored emerging trends, the potential for personalized medicine, and the ethical and social implications of this transformative technology. In conclusion, through comprehensive analysis and case studies, this paper underscores the profound impact of nanoparticles on breast cancer treatment and their future potential.
Collapse
Affiliation(s)
- Fatemah S Basingab
- Department of Biological Sciences, Faculty of Science, King Abdulaziz University, Jeddah, 21859, Saudi Arabia
- Immunology Unit, King Fahd Medical Research Center, King Abdulaziz University, Jeddah, 21859, Saudi Arabia
| | - Omniah A Alshahrani
- Department of Biological Sciences, Faculty of Science, King Abdulaziz University, Jeddah, 21859, Saudi Arabia
| | - Ibtehal H Alansari
- Department of Biological Sciences, Faculty of Science, King Abdulaziz University, Jeddah, 21859, Saudi Arabia
| | - Nada A Almarghalani
- Department of Biological Sciences, Faculty of Science, King Abdulaziz University, Jeddah, 21859, Saudi Arabia
| | - Nada H Alshelali
- Department of Biological Sciences, Faculty of Science, King Abdulaziz University, Jeddah, 21859, Saudi Arabia
| | - Abeer Hamad Alsaiary
- Biology Department, College of Sciences, Imam Abdulrahman Bin Faisal University, Dammam, Saudi Arabia
| | - Najwa Alharbi
- Department of Biology Science, Faculty of Science, King Abdulaziz University, Jeddah, 21859, Saudi Arabia
| | - Kawther A Zaher
- Immunology Unit, King Fahd Medical Research Center, King Abdulaziz University, Jeddah, 21859, Saudi Arabia
- Department of Medical Laboratory Sciences, Faculty of Applied Medical Sciences, King Abdulaziz University, Jeddah, 21859, Saudi Arabia
| |
Collapse
|
11
|
Garitaonaindia Y, Martínez-Cutillas M, Uribarren M, Redondo I, Calvo V, Serna-Blasco R, Provencio M. Adoptive cell therapies in thoracic malignancies: a comprehensive review. Clin Transl Oncol 2025:10.1007/s12094-024-03834-5. [PMID: 39789380 DOI: 10.1007/s12094-024-03834-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2024] [Accepted: 12/19/2024] [Indexed: 01/12/2025]
Abstract
This review aims to summarize recent studies and findings within adoptive cell therapies, including tumor-infiltrating lymphocytes, genetically engineered T cell receptors, and chimeric antigen receptor T cells, in the treatment of thoracic malignancies, including non-small cell lung cancer, small cell lung cancer, and malignant pleural mesothelioma. Several trials are ongoing, and a few have reported results, suggesting that adoptive cell therapies may represent a potential treatment option for these patients, especially when checkpoint inhibition has failed. We also discuss the potential implementation of these therapies, as they present a new toxicity profile and an intrinsic financial burden. Despite the challenges to overcome, such as the accurate identification of antigens and developing strategies to improve efficacy and toxicity profiles, new cellular therapies are experiencing significant development in the field of thoracic malignancies.
Collapse
Affiliation(s)
- Yago Garitaonaindia
- Medical Oncology Department, Puerta de Hierro University Hospital, C/ Manuel de Falla, 1, 28222, Majadahonda, Madrid, Spain.
| | - Marta Martínez-Cutillas
- Medical Oncology Department, Puerta de Hierro University Hospital, C/ Manuel de Falla, 1, 28222, Majadahonda, Madrid, Spain
| | - Maria Uribarren
- Medical Oncology Department, Puerta de Hierro University Hospital, C/ Manuel de Falla, 1, 28222, Majadahonda, Madrid, Spain
| | - Isabel Redondo
- Medical Oncology Department, Puerta de Hierro University Hospital, C/ Manuel de Falla, 1, 28222, Majadahonda, Madrid, Spain
| | - Virginia Calvo
- Medical Oncology Department, Puerta de Hierro University Hospital, C/ Manuel de Falla, 1, 28222, Majadahonda, Madrid, Spain
| | - Roberto Serna-Blasco
- Instituto de Investigación Sanitaria Puerta de Hierro-Segovia de Arana (IDIPHISA), Puerta De Hierro University Hospital, Majadahonda, Madrid, Spain
| | - Mariano Provencio
- Medical Oncology Department, Puerta de Hierro University Hospital, C/ Manuel de Falla, 1, 28222, Majadahonda, Madrid, Spain.
| |
Collapse
|
12
|
Avouac J, Scherlinger M. CAR T-Cell Therapy for Rheumatic Diseases: What Does the Future Hold? BioDrugs 2025; 39:5-19. [PMID: 39738985 DOI: 10.1007/s40259-024-00692-z] [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: 11/13/2024] [Indexed: 01/02/2025]
Abstract
Chimeric antigen receptor (CAR) T-cell therapy, initially successful in treating hematological malignancies, is emerging as a potential treatment for autoimmune diseases, including rheumatic conditions. CAR T cells, engineered to target and eliminate autoreactive B cells, offer a novel approach to managing diseases like systemic lupus erythematosus (SLE), systemic sclerosis (SSc), and inflammatory myopathies, where B cells play a pivotal role in disease pathology. Early case reports have demonstrated promising results, with patients achieving significant disease remission, normalization of serological markers, and the ability to discontinue traditional immunosuppressive therapies, which supported the initiation of several clinical trials. However, the application of CAR T-cell therapy in chronic inflammatory rheumatic disorders poses unique challenges, including patient heterogeneity, the risk of adverse effects such as cytokine release syndrome, and the high costs associated with the therapy. Despite these challenges, the potential for CAR T cells to provide long-term remission or even a cure in refractory autoimmune diseases is significant. Ongoing research aims to optimize CAR T-cell constructs and improve safety profiles, paving the way for broader application in rheumatic diseases. If these challenges can be addressed, CAR T-cell therapy could revolutionize the treatment landscape for chronic inflammatory rheumatic disorders, offering new hope for patients with severe, treatment-resistant conditions.
Collapse
Affiliation(s)
- Jérôme Avouac
- Service de Rhumatologie, Hôpital Cochin, AP-HP, CUP, Université Paris Descartes, 27 rue du Faubourg Saint-Jacques, 75014, Paris, France.
- INSERM U1016 and UMR8104, Institut Cochin, 75014, Paris, France.
| | - Marc Scherlinger
- Rheumatology Department, Strasbourg University Hospital, 1 Avenue Molière, 67000, Strasbourg, France
- UMR_S INSERM 1109, Immunorhumatologie moléculaire, 1 place de l'hôpital, 67000, Strasbourg, France
| |
Collapse
|
13
|
Sainatham C, Yadav D, Dilli Babu A, Tallapalli JR, Kanagala SG, Filippov E, Murillo Chavez F, Ahmed N, Lutfi F. The current socioeconomic and regulatory landscape of immune effector cell therapies. Front Med (Lausanne) 2024; 11:1462307. [PMID: 39697210 PMCID: PMC11652178 DOI: 10.3389/fmed.2024.1462307] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2024] [Accepted: 11/18/2024] [Indexed: 12/20/2024] Open
Abstract
Immune cell effector therapies, including chimeric antigen receptor (CAR)-T cells, T-cell receptor (TCR) T cells, natural killer (NK) cells, and macrophage-based therapies, represent a transformative approach to cancer treatment, harnessing the immune system to target and eradicate malignant cells. CAR-T cell therapy, the most established among these, involves engineering T cells to express CARs specific to cancer cell antigens, showing remarkable efficacy in hematologic malignancies like leukemias, B-cell lymphomas, and multiple myeloma. Similarly, TCR-modified therapies, which reprogram T cells to recognize intracellular tumor antigens presented by major histocompatibility complex (MHC) molecules, offer promise for a range of solid tumors. NK-cell therapies leverage NK cells' innate cytotoxicity, providing an allogeneic approach that avoids some of the immune-related complications associated with T-cell-based therapies. Macrophage-based therapies, still in early stages of the development, focus on reprogramming macrophages to stimulate an immune response against cancer cells in the tumor microenvironment. Despite their promise, socioeconomic and regulatory challenges hinder the accessibility and scalability of immune cell effector therapies. These treatments are costly, with CAR-T therapies currently exceeding $400,000 per patient, creating significant disparities in access based on socioeconomic status and geographic location. The high manufacturing costs stem from the personalized, labor-intensive processes of harvesting, modifying, and expanding patients' cells. Moreover, complex logistics for manufacturing and delivering these therapies limit their reach, particularly in low-resource settings. Regulatory pathways further complicate the landscape. In the United States., the Food and Drug Administrations' (FDA) accelerated approval processes for cell-based therapies facilitate innovation but do not address cost-related barriers. In Europe, the European Medicines Agency (EMA) offers adaptive pathways, yet decentralized reimbursement systems create uneven access across member states. Additionally, differing regulatory standards for manufacturing and quality control worldwide pose hurdles for global harmonization and access. To expand the reach of immune effector cell therapies, a multipronged approach is needed-streamlined regulatory frameworks, policies to reduce treatment costs, and international collaborations to standardize manufacturing. Addressing these socioeconomic and regulatory obstacles is essential to make these life-saving therapies accessible to a broader patient population worldwide. We present a literature review on the current landscape of immune effector cell therapies and barriers of access to currently approved standard of care therapy at various levels.
Collapse
Affiliation(s)
- Chiranjeevi Sainatham
- Department of Internal Medicine, Sinai Hospital of Baltimore, Baltimore, MD, United States
| | - Devvrat Yadav
- Department of Internal Medicine, Sinai Hospital of Baltimore, Baltimore, MD, United States
| | - Aravind Dilli Babu
- Department of Internal Medicine, Sinai Hospital of Baltimore, Baltimore, MD, United States
| | - Jayanth Reddy Tallapalli
- Division of Infectious Diseases, Department of Internal Medicine, University of South Florida, Tampa, FL, United States
| | - Sai Gautham Kanagala
- Department of Internal Medicine, New York Medical College/Metropolitan Hospital Center, New York, NY, United States
| | - Evgenii Filippov
- Department of Internal Medicine, Sinai Hospital of Baltimore, Baltimore, MD, United States
| | - Franco Murillo Chavez
- Department of Internal Medicine, Sinai Hospital of Baltimore, Baltimore, MD, United States
| | - Nausheen Ahmed
- Department of Hematologic Malignancies and Cellular Therapeutics, University of Kansas Medical Center, Kansas City, KS, United States
| | - Forat Lutfi
- Department of Hematologic Malignancies and Cellular Therapeutics, University of Kansas Medical Center, Kansas City, KS, United States
| |
Collapse
|
14
|
Gye A, De Abreu Lourenco R, Goodall S. Different Models, Same Results: Considerations When Choosing Between Approaches to Model Cost Effectiveness of Chimeric-Antigen Receptor T-Cell Therapy Versus Standard of Care. PHARMACOECONOMICS 2024; 42:1359-1371. [PMID: 39243347 PMCID: PMC11564325 DOI: 10.1007/s40273-024-01430-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Accepted: 08/18/2024] [Indexed: 09/09/2024]
Abstract
OBJECTIVE Chimeric antigen-receptor T-cell therapy (CAR-T) is characterised by early phase data at the time of registration, high upfront cost and a complex manufacturing and administration process compared with standard therapies. Our objective was to compare the performance of different models to assess the cost effectiveness of CAR-T using a state-transition model (STM), partitioned survival model (PSM) and discrete event simulation (DES). METHODS Individual data for tisagenlecleucel for the treatment of young patients with acute lymphoblastic leukaemia (ALL) were used to populate the models. Costs and benefits were measured over a lifetime to generate a cost per quality-adjusted life-year (QALY). Model performance was compared quantitatively on the outcomes generated and a checklist developed summarising the components captured by each model type relevant to assessing cost effectiveness of CAR-T. RESULTS Models generated similar results with base-case analyses ranging from an incremental cost per QALY of $96,074-$99,625. DES was the only model to specifically capture CAR-T wait time, demonstrating a substantial loss of benefit of CAR-T with increased wait time. CONCLUSION Although model type did not meaningfully impact base-case results, the ability to incorporate an outcome-based payment arrangement (OBA) and wait time are important elements to consider when selecting a model for CAR-T. DES provided greater flexibility compared with STM and PSM approaches to deal with the complex manufacturing and administration process that can lead to extended wait times and substantially reduce the benefit of CAR-T. This is an important consideration when selecting a model type for CAR-T, so major drivers of uncertainty are considered in funding decisions.
Collapse
Affiliation(s)
- Amy Gye
- Centre for Health Economics Research and Evaluation, University of Technology Sydney, Sydney, New South Wales, Australia.
| | - Richard De Abreu Lourenco
- Centre for Health Economics Research and Evaluation, University of Technology Sydney, Sydney, New South Wales, Australia
| | - Stephen Goodall
- Centre for Health Economics Research and Evaluation, University of Technology Sydney, Sydney, New South Wales, Australia
| |
Collapse
|
15
|
Kato K, Nakamura S, Wakana A, Koh Y, Izutsu K. Pembrolizumab in Japanese patients with primary mediastinal large B-cell lymphoma: results from the KEYNOTE-A33 study. Int J Clin Oncol 2024; 29:1977-1983. [PMID: 39294486 DOI: 10.1007/s10147-024-02627-8] [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: 07/15/2024] [Accepted: 09/12/2024] [Indexed: 09/20/2024]
Abstract
BACKGROUND KEYNOTE-A33 (NCT04317066) is an open-label, single-arm, phase 1 trial designed to evaluate the safety and efficacy of pembrolizumab in Japanese patients with relapsed or refractory (R/R) primary mediastinal large B-cell lymphoma (PMBCL). METHODS Patients received pembrolizumab 200 mg every 3 weeks for up to 35 cycles. The primary endpoints were safety and objective response rate (ORR) per International Working Group 2007 criteria by independent central review. The secondary endpoint was disease control rate (DCR). Duration of response (DOR), progression-free survival (PFS), and overall survival (OS) were exploratory. RESULTS Seven patients were enrolled and treated; the median age was 32 years (range 26-43) and 86% were female. The median time from the first dose to data cutoff (April 12, 2022) was 5.6 months (range 2.4-21.2). Grade 3-5 treatment-related adverse events (AEs) occurred in 2 patients (29%; 2 grade 4 neutropenia, 1 grade 3 febrile neutropenia); however, no patient discontinued pembrolizumab or died because of treatment-related AEs. The ORR was 43% [95% confidence interval (CI) 10-82]. DCR was 57% (95% CI 18-90). Median DOR was not reached (NR). Four (57%) patients had a reduction in target lesion size of ≥ 50%. The median PFS was 2.9 months (95% CI 2.6-NR). The median OS was 17.5 months (95% CI NE-NE), and the 12 months OS rate was 100%. CONCLUSION Overall, pembrolizumab had manageable safety and clinically meaningful antitumor activity in Japanese patients with R/R PMBCL, results that were consistent with those observed in prior global studies. TRIAL REGISTRY Registry and the Registration No. of the study/trial: Clinicaltrials.gov: NCT04317066.
Collapse
MESH Headings
- Humans
- Antibodies, Monoclonal, Humanized/therapeutic use
- Antibodies, Monoclonal, Humanized/adverse effects
- Female
- Male
- Adult
- Lymphoma, Large B-Cell, Diffuse/drug therapy
- Lymphoma, Large B-Cell, Diffuse/pathology
- Mediastinal Neoplasms/drug therapy
- Mediastinal Neoplasms/pathology
- Mediastinal Neoplasms/mortality
- Antineoplastic Agents, Immunological/therapeutic use
- Antineoplastic Agents, Immunological/adverse effects
- Japan
- Progression-Free Survival
- East Asian People
Collapse
Affiliation(s)
- Koji Kato
- Department of Medicine and Biosystemic Science, Kyushu University Graduate School of Medical Sciences, 3-1-1 Maidashi, Higashi-Ku, Fukuoka, 812-8582, Japan.
| | - Sosuke Nakamura
- MSD.K.K., 1-13-12 Kudan-Kita, Chiyoda-Ku, Tokyo, 102-0073, Japan
| | - Akira Wakana
- MSD.K.K., 1-13-12 Kudan-Kita, Chiyoda-Ku, Tokyo, 102-0073, Japan
| | - Yasuhiro Koh
- MSD.K.K., 1-13-12 Kudan-Kita, Chiyoda-Ku, Tokyo, 102-0073, Japan
| | - Koji Izutsu
- National Cancer Center Hospital, 5-1-1 Tsukiji, Chuo-Ku, Tokyo, 104-0045, Japan
| |
Collapse
|
16
|
Crombie JL, Jun M, Wang T, Mutebi A, Wang A, Chhibber A, Kamalakar R, Ukropec J, Blaedel J, Kalsekar A. Real-world outcomes with novel therapies in relapsed/refractory diffuse large B-cell lymphoma. Leuk Lymphoma 2024; 65:1623-1633. [PMID: 39011744 DOI: 10.1080/10428194.2024.2371472] [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: 12/20/2023] [Revised: 05/31/2024] [Accepted: 06/17/2024] [Indexed: 07/17/2024]
Abstract
This study used COTA de-identified data (2010-2021) of patients in the US to explore outcomes of novel therapies in relapsed/refractory (R/R) diffuse large B-cell lymphoma (DLBCL) in real-world settings. Demographics, clinical characteristics, and clinical outcomes of patients with R/R DLBCL who received novel treatments including chimeric antigen receptor T-cell (CAR T) therapy and tafasitamab- or polatuzumab-based therapies were evaluated. Overall, 175 patients with R/R DLBCL were analyzed; 73, 69, and 27 received CAR T therapy, polatuzumab-based regimens, and tafasitamab-based regimens, respectively. In patients who had ≥1 prior lines of therapy (i.e. starting second-line or later therapy; 2 L+), CAR T, polatuzumab-based regimens, and tafasitamab-based regimens achieved a median overall survival of 26.5, 7.8, and 6.3 months, respectively. Outcomes were particularly poor for patients with relapse following CAR T, indicating that polatuzumab- and tafasitamab-based regimens in 2 L + R/R DLBCL have suboptimal outcomes in the real world. Additional treatment options are needed.
Collapse
MESH Headings
- Humans
- Lymphoma, Large B-Cell, Diffuse/drug therapy
- Lymphoma, Large B-Cell, Diffuse/mortality
- Lymphoma, Large B-Cell, Diffuse/therapy
- Lymphoma, Large B-Cell, Diffuse/pathology
- Male
- Female
- Middle Aged
- Aged
- Adult
- Immunotherapy, Adoptive/methods
- Neoplasm Recurrence, Local/pathology
- Antineoplastic Combined Chemotherapy Protocols/therapeutic use
- Antineoplastic Combined Chemotherapy Protocols/adverse effects
- Treatment Outcome
- Aged, 80 and over
- Drug Resistance, Neoplasm
- Young Adult
- Receptors, Chimeric Antigen
- Antibodies, Monoclonal, Humanized/therapeutic use
- Salvage Therapy/methods
- Retrospective Studies
- Combined Modality Therapy
Collapse
|
17
|
Garg P, Pareek S, Kulkarni P, Horne D, Salgia R, Singhal SS. Next-Generation Immunotherapy: Advancing Clinical Applications in Cancer Treatment. J Clin Med 2024; 13:6537. [PMID: 39518676 PMCID: PMC11546714 DOI: 10.3390/jcm13216537] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2024] [Revised: 10/28/2024] [Accepted: 10/28/2024] [Indexed: 11/16/2024] Open
Abstract
Next-generation immunotherapies have revolutionized cancer treatment, offering hope for patients with hard-to-treat tumors. This review focuses on the clinical applications and advancements of key immune-based therapies, including immune checkpoint inhibitors, CAR-T cell therapy, and new cancer vaccines designed to harness the immune system to combat malignancies. A prime example is the success of pembrolizumab in the treatment of advanced melanoma, underscoring the transformative impact of these therapies. Combination treatments, integrating immunotherapy with chemotherapy, radiation, and targeted therapies, are demonstrating synergistic benefits and improving patient outcomes. This review also explores the evolving role of personalized immunotherapy, guided by biomarkers, genomic data, and the tumor environment, to better target individual tumors. Although significant progress has been made, challenges such as resistance, side effects, and high treatment costs persist. Technological innovations, including nanotechnology and artificial intelligence, are explored as future enablers of these therapies. The review evaluates key clinical trials, breakthroughs, and the emerging immune-modulating agents and advanced delivery systems that hold great promise for enhancing treatment efficacy, reducing toxicity, and expanding access to immunotherapy. In conclusion, this review highlights the ongoing advancements in immunotherapy that are reshaping cancer care, with future strategies poised to overcome current challenges and further extend therapeutic reach.
Collapse
Affiliation(s)
- Pankaj Garg
- Department of Chemistry, GLA University, Mathura 281406, Uttar Pradesh, India
| | - Siddhika Pareek
- Departments of Medical Oncology & Therapeutics Research, Beckman Research Institute of City of Hope, Comprehensive Cancer Center and National Medical Center, Duarte, CA 91010, USA
| | - Prakash Kulkarni
- Departments of Medical Oncology & Therapeutics Research, Beckman Research Institute of City of Hope, Comprehensive Cancer Center and National Medical Center, Duarte, CA 91010, USA
| | - David Horne
- Departments of Molecular Medicine, Beckman Research Institute of City of Hope, Comprehensive Cancer Center and National Medical Center, Duarte, CA 91010, USA
| | - Ravi Salgia
- Departments of Medical Oncology & Therapeutics Research, Beckman Research Institute of City of Hope, Comprehensive Cancer Center and National Medical Center, Duarte, CA 91010, USA
| | - Sharad S. Singhal
- Departments of Medical Oncology & Therapeutics Research, Beckman Research Institute of City of Hope, Comprehensive Cancer Center and National Medical Center, Duarte, CA 91010, USA
| |
Collapse
|
18
|
Blud D, Rubio-Reyes P, Perret R, Weinkove R. Tuning CAR T-cell therapies for efficacy and reduced toxicity. Semin Hematol 2024; 61:333-344. [PMID: 39095226 DOI: 10.1053/j.seminhematol.2024.07.003] [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: 05/30/2024] [Revised: 06/24/2024] [Accepted: 07/03/2024] [Indexed: 08/04/2024]
Abstract
Chimeric antigen receptor (CAR) T-cell therapies are a standard of care for certain relapsed or refractory B-cell cancers. However, many patients do not respond to CAR T-cell therapy or relapse later, short- and long-term toxicities are common, and current CAR T-cell therapies have limited efficacy for solid cancers. The gene engineering inherent in CAR T-cell manufacture offers an unprecedented opportunity to control cellular characteristics and design products that may overcome these limitations. This review summarises available methods to "tune" CAR T-cells for optimal efficacy and safety. The components of a typical CAR, and the modifications that can influence CAR T-cell function are discussed. Methods of engineering passive, inducible or autonomous control mechanisms into CAR T-cells, allowing selective limitation or enhancement of CAR T-cell activity are reviewed. The impact of manufacturing processes on CAR T-cell function are considered, including methods of limiting CAR T-cell terminal differentiation and exhaustion, and the use of specific T-cell subsets as the CAR T starting material. We discuss the use of multicistronic transgenes and multiplexed gene editing. Finally, we highlight the need for innovative clinical trial designs if we are to make the most of the opportunities offered by CAR T-cell therapies.
Collapse
Affiliation(s)
- Danielle Blud
- Cancer Immunotherapy Programme, Malaghan Institute of Medical Research, Wellington, New Zealand
| | - Patricia Rubio-Reyes
- Cancer Immunotherapy Programme, Malaghan Institute of Medical Research, Wellington, New Zealand
| | - Rachel Perret
- Cancer Immunotherapy Programme, Malaghan Institute of Medical Research, Wellington, New Zealand
| | - Robert Weinkove
- Cancer Immunotherapy Programme, Malaghan Institute of Medical Research, Wellington, New Zealand; Wellington Blood & Cancer Centre, Te Whatu Ora Health New Zealand Capital Coast & Hutt Valley, Wellington, New Zealand; Department of Pathology and Molecular Medicine, University of Otago Wellington, Wellington, New Zealand; Maurice Wilkins Centre for Molecular Biodiscovery, Auckland, New Zealand.
| |
Collapse
|
19
|
Siefen AC, Kurte MS, Jakobs F, Teichert M, von Tresckow B, Reinhardt HC, Holtick U, Atta J, Jehn C, Sala E, Warnecke A, Hänel M, Scheid C, Kron F. How to optimize the CAR-T Cell therapy process? A group concept mapping analysis of preconditions for a frictionless process from a German multistakeholder perspective. Front Oncol 2024; 14:1466803. [PMID: 39376984 PMCID: PMC11456517 DOI: 10.3389/fonc.2024.1466803] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2024] [Accepted: 08/28/2024] [Indexed: 10/09/2024] Open
Abstract
Introduction Treatment with chimeric antigen receptor T (CAR-T) cells involves a large number of interdisciplinary stakeholders and is associated with complex processes ranging from patient-specific production to follow-up care. Due to the complexity, maximum process optimization is required in order to avoid efficiency losses. This study aimed at systematically determining the preconditions for a frictionless flow of the CAR-T process by surveying the stakeholders involved. Methods A Group Concept Mapping (GCM) analysis, a mixed-methods participatory research, was conducted. CAR-T experts from different professional backgrounds went through three steps: 1) Brainstorming relevant aspects (statements) for a frictionless process, 2) Sorting the collected statements based on their similarity, and 3) Rating the importance and feasibility of each statement. A cluster map reflecting the overarching topics was derived, and mean ratings per statement and cluster were calculated. Results Overall, 20 CAR-T experts participated. A total of 80 statements were collected, resulting in a map of the following 10 clusters (mean importance/feasibility): Information for patients and physicians (4.16/3.77), Supportive network (4.03/3.53), Eligibility of patients (4.41/3.63), Evidence, transparency and communication (4.01/3.33), Paperwork (4.1/2.52), Interface with pharmaceutical manufacturer (4.03/2.85), Reimbursement (4.29/2.31), Quality Management (4.17/3.18), Infrastructure of CAR-T clinics (4.1/2.93), and Patient-oriented processes (4.46/3.32). Discussion The 80 statements underlined the complex and manifold nature of the CAR-T treatment process. Our results reflect the first step in overcoming hurdles: identifying potential hurdles and required preconditions. Decision-makers and stakeholders can use the results to derive strategies and measures to further promote a frictionless process.
Collapse
Affiliation(s)
- Ann-Cathrine Siefen
- Faculty of Medicine, University of Duisburg-Essen, Essen, Germany
- VITIS Healthcare Group, Cologne, Germany
| | - Melina Sophie Kurte
- Faculty of Medicine, University of Duisburg-Essen, Essen, Germany
- VITIS Healthcare Group, Cologne, Germany
| | - Florian Jakobs
- Department of Hematology and Stem Cell Transplantation, Faculty of Medicine and University Hospital Essen, University of Duisburg-Essen, Essen, Germany
| | - Marcel Teichert
- Department of Hematology and Stem Cell Transplantation, Faculty of Medicine and University Hospital Essen, University of Duisburg-Essen, Essen, Germany
| | - Bastian von Tresckow
- Department of Hematology and Stem Cell Transplantation, West German Cancer Center and German Cancer Consortium (DKTK Partner Site Essen), University Hospital Essen, University of Duisburg-Essen, Essen, Germany
| | - Hans Christian Reinhardt
- Department of Hematology and Stem Cell Transplantation, Faculty of Medicine and University Hospital Essen, University of Duisburg-Essen, Essen, Germany
| | - Udo Holtick
- Department I of Internal Medicine, Faculty of Medicine and University Hospital Cologne, University of Cologne, Cologne, Germany
| | - Johannes Atta
- Department of Hematology, Giessen and Marburg University Hospital (UKGM), Giessen, Germany
| | - Christian Jehn
- Department of Hematology/Oncology and Stem-cell Transplantation, St Georg, Asklepios Hamburg, Hamburg, Germany
| | - Elisa Sala
- Department of Internal Medicine III, University Hospital of Ulm, Ulm, Germany
| | - Anke Warnecke
- Department of Hematology and Medical Oncology, Division for Stem Cell Transplantation and Cellular Therapy, University Hospital Göttingen, Göttingen, Germany
| | - Mathias Hänel
- Department of Internal Medicine III, Klinikum Chemnitz, Chemnitz, Germany
| | - Christof Scheid
- Department I of Internal Medicine, Faculty of Medicine and University Hospital Cologne, University of Cologne, Cologne, Germany
| | - Florian Kron
- VITIS Healthcare Group, Cologne, Germany
- Department I of Internal Medicine, Faculty of Medicine and University Hospital Cologne, University of Cologne, Cologne, Germany
- Fachhochschule für Oekonomie & Management (FOM) University of Applied Sciences, Essen, Germany
| |
Collapse
|
20
|
Mishra HK, Kalyuzhny A. Revolutionizing Cancer Treatments through Stem Cell-Derived CAR T Cells for Immunotherapy: Opening New Horizons for the Future of Oncology. Cells 2024; 13:1516. [PMID: 39329700 PMCID: PMC11430090 DOI: 10.3390/cells13181516] [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: 08/13/2024] [Revised: 09/06/2024] [Accepted: 09/07/2024] [Indexed: 09/28/2024] Open
Abstract
Recent advances in cellular therapies have paved the way for innovative treatments of various cancers and autoimmune disorders. Induced pluripotent stem cells (iPSCs) represent a remarkable breakthrough, offering the potential to generate patient-specific cell types for personalized as well as allogeneic therapies. This review explores the application of iPSC-derived chimeric antigen receptor (CAR) T cells, a cutting-edge approach in allogeneic cancer immunotherapies. CAR T cells are genetically engineered immune cells designed to target specific tumor antigens, and their integration with iPSC technology holds immense promise for enhancing the efficacy, safety, and scalability of cellular therapies. This review begins by elucidating the principles behind iPSC generation and differentiation into T cells, highlighting the advantage of iPSCs in providing a uniform, inexhaustible source of CAR T cells. Additionally, we discuss the genetic modification of iPSC-derived T cells to express various CARs, emphasizing the precision and flexibility this affords in designing customized therapies for a diverse range of malignancies. Notably, iPSC-derived CAR T cells demonstrate a superior proliferative capacity, persistence, and anti-tumor activity compared to their conventionally derived counterparts, offering a potential solution to challenges associated with conventional CAR T cell therapies. In conclusion, iPSC-derived CAR T cells represent a groundbreaking advancement in cellular therapies, demonstrating unparalleled potential in revolutionizing the landscape of immunotherapies. As this technology continues to evolve, it holds the promise of providing safer, more effective, and widely accessible treatment options for patients battling cancer and other immune-related disorders. This review aims to shed light on the transformative potential of iPSC-derived CAR T cells and inspire further research and development in this dynamic field.
Collapse
|
21
|
Hungria V, Sureda A, Campelo GR, Salvino MA, Ramasamy K. Proceedings from the First Onco Summit: LATAM Chapter, 19-20 May 2023, Rio de Janeiro, Brazil. Cancers (Basel) 2024; 16:3063. [PMID: 39272921 PMCID: PMC11394439 DOI: 10.3390/cancers16173063] [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: 07/08/2024] [Revised: 08/20/2024] [Accepted: 08/28/2024] [Indexed: 09/15/2024] Open
Abstract
The Onco Summit 2023: The Latin American (LATAM) Chapter took place over two days, from 19-20 May 2023, in Brazil. The event aimed to share the latest updates across various oncology disciplines, address critical clinical challenges, and exchange best practices to ensure optimal patient treatment. More than 30 international and regional speakers and more than 300 oncology specialists participated in the Summit. The Summit discussions centered on common challenges and therapeutic advances in cancer care, with a specific focus on the unique obstacles faced in LATAM and examples of adaptable strategies to address these challenges. The Summit also facilitated the establishment of a network of oncologists, hematologists, and scientists in LATAM, enabling collaboration to improve cancer care, both in this region and globally, through drug development and clinical research. This report summarizes the key discussions from the Summit for the global and LATAM oncology community.
Collapse
Affiliation(s)
- Vania Hungria
- Hematology, Faculty of Medical Sciences of Santa Casa de São Paulo, São Paulo 01224-001, Brazil
| | - Anna Sureda
- Clinical Hematology Department, Catalan Institut Català d'Oncologia-L'Hospitalet, Instituto de Investigación Biomédica de Bellvitge (IDIBELL), University of Barcelona (UB), 08908 Barcelona, Spain
| | - Garcia Rosario Campelo
- Thoracic Tumors Unit, Medical Oncology Department, University Hospital A Coruña Biomedical Research Institute (INIBIC), 15006 A Coruña, Spain
| | - Marco Aurélio Salvino
- Cell Therapy, D'OR Institute Research & Education (IDOR)/PPGMS-Federal University of Bahia (UFBA), Salvador 40110-100, Brazil
| | - Karthik Ramasamy
- Oxford Translational Myeloma Centre, NDORMS, University of Oxford, Oxford OX3 7LD, UK
| |
Collapse
|
22
|
Atallah R, Ahmed N, Ayoobkhan F, Saif MSI, Logan E, Shrestha A, Anwer F, Mahmoudjafari Z, Mushtaq MU, Hashmi H, Ganguly S, McGuirk J, Shebli A, Abdallah AO, Banerjee R, Alkharabsheh O. TACTUM: Trends in Access to Cellular Therapies in Multiple Myeloma, Perspectives of Treating Versus Referring Physicians. Transplant Cell Ther 2024; 30:925.e1-925.e6. [PMID: 38763416 DOI: 10.1016/j.jtct.2024.05.011] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/29/2024] [Revised: 04/29/2024] [Accepted: 05/12/2024] [Indexed: 05/21/2024]
Abstract
Chimeric antigen receptor T cell therapy (CAR-T) and bispecific T cell engagers (TCE) for multiple myeloma (MM) are readily available at many large US medical centers. However, many potentially eligible patients may not be referred to the specialized centers administering these therapies. Perspectives regarding potential barriers for MM cellular therapy from referring-center oncologists (ROs) versus treating-center oncologists (TOs) have not been reported previously. We conducted TACTUM-23, a survey of US oncologists who treat MM, to identify perceived barriers to these cellular therapies. This 24-question survey, which focused on demographics and perceived barriers to CAR-T and TCE, was conducted between June and August 2023. Of 247 oncologists, 37 (15%) completed the survey including 26 (70%) TOs who prescribed both CAR-T and TCEs, 4 (11%) TOs who only prescribed TCEs, and 7 (19%) ROs who referred patients. The top RO-stated barrier to CAR-T was financial toxicity, while the top TO-stated barrier to CAR-T was leukapheresis/ manufacturing slot availability. The top RO-stated barrier to TCE was financial toxicity, while the top TO-stated barrier to TCE was the hospitalization requirement. In conclusion, financial concerns are perceived by ROs to be the top barrier to both CAR-T and TCEs in myeloma. In contrast, TOs perceive logistical concerns to be the top barrier. Interventions to lower financial toxicity during these therapies, and outreach to raise awareness of such interventions among ROs, are needed alongside strategies to streamline manufacturing (for CAR-T) and monitoring.
Collapse
Affiliation(s)
- Rawan Atallah
- Division of Hematologic Malignancies & Cellular Therapeutics, University of Kansas Medical Center, Westwood, Kansas; US Myeloma Innovations Research Collaborative (USMIRC), Westwood, Kansas.
| | - Nausheen Ahmed
- Division of Hematologic Malignancies & Cellular Therapeutics, University of Kansas Medical Center, Westwood, Kansas; US Myeloma Innovations Research Collaborative (USMIRC), Westwood, Kansas
| | - Fathima Ayoobkhan
- Department of Internal Medicine, Trinity Health Oakland/ Wayne State University, Pontiac, Michigan; US Myeloma Innovations Research Collaborative (USMIRC), Westwood, Kansas
| | - Md Saiful Islam Saif
- Department of Biostatistics and Data Science, University of Kansas Medical Center, Kansas City, Kansas
| | - Emerson Logan
- Division of Hematologic Malignancies & Cellular Therapeutics, University of Kansas Medical Center, Westwood, Kansas; US Myeloma Innovations Research Collaborative (USMIRC), Westwood, Kansas
| | - Anuj Shrestha
- Section of Hematology and Oncology, University of Missouri, Kansas City, Missouri
| | - Faiz Anwer
- Department of Hematology and Medical Oncology, Taussig Cancer Institute, Cleveland Clinic, Cleveland, Ohio; US Myeloma Innovations Research Collaborative (USMIRC), Westwood, Kansas
| | - Zahra Mahmoudjafari
- Department of Pharmacy, University of Kansas Cancer Center, Kansas City, Kansas; US Myeloma Innovations Research Collaborative (USMIRC), Westwood, Kansas
| | - Muhammed Umair Mushtaq
- Division of Hematologic Malignancies & Cellular Therapeutics, University of Kansas Medical Center, Westwood, Kansas; US Myeloma Innovations Research Collaborative (USMIRC), Westwood, Kansas
| | - Hamza Hashmi
- Department of Medicine, Multiple Myeloma service, Memorial Sloan Kettering Cancer Center, New York City, New York; US Myeloma Innovations Research Collaborative (USMIRC), Westwood, Kansas
| | | | - Joseph McGuirk
- Division of Hematologic Malignancies & Cellular Therapeutics, University of Kansas Medical Center, Westwood, Kansas
| | - Atrash Shebli
- Department of Hematologic Oncology & Blood Disorders, Atrium Health Levine Cancer Institute, Charlotte, North Carolina; US Myeloma Innovations Research Collaborative (USMIRC), Westwood, Kansas
| | - Al-Ola Abdallah
- Division of Hematologic Malignancies & Cellular Therapeutics, University of Kansas Medical Center, Westwood, Kansas; US Myeloma Innovations Research Collaborative (USMIRC), Westwood, Kansas
| | | | - Omar Alkharabsheh
- University of South Alabama, Mitchell Cancer Institute, Mobile, Alabama; US Myeloma Innovations Research Collaborative (USMIRC), Westwood, Kansas
| |
Collapse
|
23
|
Holland SM, Sohal A, Nand AA, Hutmacher DW. A quest for stakeholder synchronization in the CAR T-cell therapy supply chain. Front Bioeng Biotechnol 2024; 12:1413688. [PMID: 39175619 PMCID: PMC11338886 DOI: 10.3389/fbioe.2024.1413688] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2024] [Accepted: 07/22/2024] [Indexed: 08/24/2024] Open
Abstract
Advancements in cell therapy have the potential to improve healthcare accessibility for eligible patients. However, there are still challenges in scaling production and reducing costs. These challenges involve various stakeholders such as the manufacturing facility, third-party logistics (3PL) company, and medical center. Proposed solutions tend to focus on individual companies rather than addressing the interconnectedness of the supply chain's challenges. The challenges can be categorized as barriers from product characteristics, regulatory requirements, or lagging infrastructure. Each barrier affects multiple stakeholders, especially during a boundary event like product handover. Therefore, solutions that only consider the objectives of one stakeholder fail to address underlying problems. This review examines the interconnecting cell therapy supply chain challenges and how they affect the multiple stakeholders involved. The authors consider whether proposed solutions impact individual stakeholders or the entire supply chain and discuss the benefits of stakeholder coordination-focused solutions such as integrated technologies and information tracking. The review highlights how coordination efforts allow for the implementation of widely-supported cell therapy supply solutions such as decentralized manufacturing through stakeholder collaboration.
Collapse
Affiliation(s)
- Shelby M. Holland
- Department of Management, Monash Business School, Monash University Caufield Campus, Melbourne, VIC, Australia
- Australian Research Council Training Centre for Cell and Tissue Engineering Technologies, Monash University Clayton Campus, Melbourne, VIC, Australia
| | - Amrik Sohal
- Department of Management, Monash Business School, Monash University Caufield Campus, Melbourne, VIC, Australia
- Australian Research Council Training Centre for Cell and Tissue Engineering Technologies, Monash University Clayton Campus, Melbourne, VIC, Australia
| | - Alka Ashwini Nand
- Department of Management, Monash Business School, Monash University Caufield Campus, Melbourne, VIC, Australia
- Australian Research Council Training Centre for Cell and Tissue Engineering Technologies, Monash University Clayton Campus, Melbourne, VIC, Australia
| | - Dietmar W. Hutmacher
- Australian Research Council Training Centre for Cell and Tissue Engineering Technologies, Monash University Clayton Campus, Melbourne, VIC, Australia
- Faculty of Engineering, School of Mechanical Medical and Process Engineering, Queensland University of Technology, Brisbane, QLD, Australia
- Australian Research Council Training Centre for Multiscale 3D Imaging, Modelling and Manufacturing (M3D Innovation), Queensland University of Technology, Kelvin Grove, QLD, Australia
- Max Planck Queensland Centre, Queensland University of Technology, Brisbane, QLD, Australia
| |
Collapse
|
24
|
Srivastava S, Singh S, Singh A. Augmenting the landscape of chimeric antigen receptor T-cell therapy. Expert Rev Anticancer Ther 2024; 24:755-773. [PMID: 38912754 DOI: 10.1080/14737140.2024.2372330] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2024] [Accepted: 06/21/2024] [Indexed: 06/25/2024]
Abstract
INTRODUCTION The inception of recombinant DNA technology and live cell genomic alteration have paved the path for the excellence of cell and gene therapies and often provided the first curative treatment for many indications. The approval of the first Chimeric Antigen Receptor (CAR) T-cell therapy was one of the breakthrough innovations that became the headline in 2017. Currently, the therapy is primarily restricted to a few nations, and the market is growing at a CAGR (current annual growth rate) of 11.6% (2022-2032), as opposed to the established bio-therapeutic market at a CAGR of 15.9% (2023-2030). The limited technology democratization is attributed to its autologous nature, lack of awareness, therapy inclusion criteria, high infrastructure cost, trained personnel, complex manufacturing processes, regulatory challenges, recurrence of the disease, and long-term follow-ups. AREAS COVERED This review discusses the vision and strategies focusing on the CAR T-cell therapy democratization with mitigation plans. Further, it also covers the strategies to leverage the mRNA-based CAR T platform for building an ecosystem to ensure availability, accessibility, and affordability to the community. EXPERT OPINION mRNA-guided CAR T cell therapy is a rapidly growing area wherein a collaborative approach among the stakeholders is needed for its success.
Collapse
Affiliation(s)
| | - Sanjay Singh
- mRNA Department, Gennova Biopharmaceuticals Ltd. ITBT Park, Pune, India
| | - Ajay Singh
- mRNA Department, Gennova Biopharmaceuticals Ltd. ITBT Park, Pune, India
| |
Collapse
|
25
|
Medina-Olivares FJ, Gómez-De León A, Ghosh N. Obstacles to global implementation of CAR T cell therapy in myeloma and lymphoma. Front Oncol 2024; 14:1397613. [PMID: 39099684 PMCID: PMC11294242 DOI: 10.3389/fonc.2024.1397613] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2024] [Accepted: 07/01/2024] [Indexed: 08/06/2024] Open
Abstract
Chimeric Antigen Receptor T-cell (CAR-T) therapies are transforming the treatment of B-cell lymphoproliferative disorders and multiple myeloma, yet global access challenges and barriers for their implementation persist. Global access disparities persist, particularly for persons living in low and middle-income countries and for underserved populations in high income countries. In this review we address patient-related factors including age, comorbidities, fitness, race and ethnicity, and geographic location for CAR-T access. Also, we review disease-related and health system barriers like disease biology, potential for short and long-term toxicity, insurance access, referrals, supply and manufacturing, regulation, costs and treatment center capacity. Lastly, alternatives for overcoming these barriers exemplified by research efforts worldwide are discussed, emphasizing the need for a multifaceted approach from all stakeholders to improve global accessibility and ensure equitable access and improved outcomes for patients worldwide.
Collapse
Affiliation(s)
- Fernando J. Medina-Olivares
- Facultad de Medicina y Hospital Universitario Dr. Jose Eleuterio Gonzalez, Universidad Autonoma de Nuevo Leon, Monterrey, Mexico
| | - Andrés Gómez-De León
- Facultad de Medicina y Hospital Universitario Dr. Jose Eleuterio Gonzalez, Universidad Autonoma de Nuevo Leon, Monterrey, Mexico
| | - Nilanjan Ghosh
- Atrium Health Levine Cancer Institute, Wake Forest School of Medicine, Charlotte, NC, United States
| |
Collapse
|
26
|
Issa II, Due H, Brøndum RF, Veeravakaran V, Haraldsdóttir H, Sylvester C, Brogaard A, Dhanjal S, Schmierer B, Dybkær K. CRISPR-Cas9 Knockout Screens Identify DNA Damage Response Pathways and BTK as Essential for Cisplatin Response in Diffuse Large B-Cell Lymphoma. Cancers (Basel) 2024; 16:2437. [PMID: 39001501 PMCID: PMC11240649 DOI: 10.3390/cancers16132437] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2024] [Revised: 06/27/2024] [Accepted: 06/28/2024] [Indexed: 07/16/2024] Open
Abstract
The recurrence of diffuse large B-cell lymphoma (DLBCL) has been observed in 40% of cases. The standard of care for refractory/relapsed DLBCL (RR-DLBCL) is platinum-based treatment prior to autologous stem cell transplantation; however, the prognosis for RR-DLBCL patients remains poor. Thus, to identify genes affecting the cisplatin response in DLBCL, cisplatin-based whole-genome CRISPR-Cas9 knockout screens were performed in this study. We discovered DNA damage response (DDR) pathways as enriched among identified sensitizing CRISPR-mediated gene knockouts. In line, the knockout of the nucleotide excision repair genes XPA and ERCC6 sensitized DLBCL cells to platinum drugs irrespective of proliferation rate, thus documenting DDR as essential for cisplatin sensitivity in DLBCL. Functional analysis revealed that the loss of XPA and ERCC6 increased DNA damage levels and altered cell cycle distribution. Interestingly, we also identified BTK, which is involved in B-cell receptor signaling, to affect cisplatin response. The knockout of BTK increased cisplatin sensitivity in DLBCL cells, and combinatory drug screens revealed a synergistic effect of the BTK inhibitor, ibrutinib, with platinum drugs at low concentrations. Applying local and external DLBCL cohorts, we addressed the clinical relevance of the genes identified in the CRISPR screens. BTK was among the most frequently mutated genes with a frequency of 3-5%, and XPA and ERCC6 were also mutated, albeit at lower frequencies. Furthermore, 27-54% of diagnostic DLBCL samples had mutations in pathways that can sensitize cells to cisplatin. In conclusion, this study shows that XPA and ERCC6, in addition to BTK, are essential for the response to platinum-based drugs in DLBCL.
Collapse
Affiliation(s)
- Issa Ismail Issa
- Department of Hematology, Clinical Cancer Research Center, Aalborg University Hospital, 9000 Aalborg, Denmark
- Department of Clinical Medicine, Aalborg University, 9000 Aalborg, Denmark
| | - Hanne Due
- Department of Hematology, Clinical Cancer Research Center, Aalborg University Hospital, 9000 Aalborg, Denmark
| | - Rasmus Froberg Brøndum
- Center for Clinical Data Science (CLINDA), Department of Clinical Medicine, Aalborg University, and Research, Education and Innovation, Aalborg University Hospital, 9260 Gistrup, Denmark
| | - Vidthdyan Veeravakaran
- Department of Hematology, Clinical Cancer Research Center, Aalborg University Hospital, 9000 Aalborg, Denmark
| | - Hulda Haraldsdóttir
- Department of Hematology, Clinical Cancer Research Center, Aalborg University Hospital, 9000 Aalborg, Denmark
| | - Cathrine Sylvester
- Department of Hematology, Clinical Cancer Research Center, Aalborg University Hospital, 9000 Aalborg, Denmark
| | - Asta Brogaard
- Department of Hematology, Clinical Cancer Research Center, Aalborg University Hospital, 9000 Aalborg, Denmark
| | - Soniya Dhanjal
- CRISPR Functional Genomics, SciLifeLab and Karolinska Institutet, Department of Medical Biochemistry and Biophysics, 17165 Solna, Sweden
| | - Bernhard Schmierer
- CRISPR Functional Genomics, SciLifeLab and Karolinska Institutet, Department of Medical Biochemistry and Biophysics, 17165 Solna, Sweden
| | - Karen Dybkær
- Department of Hematology, Clinical Cancer Research Center, Aalborg University Hospital, 9000 Aalborg, Denmark
- Department of Clinical Medicine, Aalborg University, 9000 Aalborg, Denmark
| |
Collapse
|
27
|
Lee NK, Chang JW. Manufacturing Cell and Gene Therapies: Challenges in Clinical Translation. Ann Lab Med 2024; 44:314-323. [PMID: 38361427 PMCID: PMC10961620 DOI: 10.3343/alm.2023.0382] [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: 09/26/2023] [Revised: 11/24/2023] [Accepted: 01/29/2024] [Indexed: 02/17/2024] Open
Abstract
The safety and efficacy of both cell and gene therapies have been demonstrated in numerous preclinical and clinical trials. Chimeric antigen receptor T (CAR-T) cell therapy, which leverages the technologies of both cell and gene therapies, has also shown great promise for treating various cancers. Advancements in pertinent fields have also highlighted challenges faced while manufacturing cell and gene therapy products. Potential problems and obstacles must be addressed to ease the clinical translation of individual therapies. Literature reviews of representative cell-based, gene-based, and cell-based gene therapies with regard to their general manufacturing processes, the challenges faced during manufacturing, and QC specifications are limited. We review the general manufacturing processes of cell and gene therapies, including those involving mesenchymal stem cells, viral vectors, and CAR-T cells. The complexities associated with the manufacturing processes and subsequent QC/validation processes may present challenges that could impede the clinical progression of the products. This article addresses these potential challenges. Further, we discuss the use of the manufacturing model and its impact on cell and gene therapy.
Collapse
Affiliation(s)
- Na Kyung Lee
- Department of Health Sciences and Technology, Samsung Advanced Institute for Health Sciences and Technology (SAIHST), Sungkyunkwan University, Seoul, Korea
- Cell and Gene Therapy Institute (CGTI), Research Institute for Future Medicine, Samsung Medical Center, Seoul, Korea
| | - Jong Wook Chang
- Department of Health Sciences and Technology, Samsung Advanced Institute for Health Sciences and Technology (SAIHST), Sungkyunkwan University, Seoul, Korea
- Cell and Gene Therapy Institute (CGTI), Research Institute for Future Medicine, Samsung Medical Center, Seoul, Korea
- Cell and Gene Therapy Institute, ENCell Co. Ltd., Seoul, Korea
| |
Collapse
|
28
|
Cui C, Feng C, Rosenthal N, Wade SW, Curry L, Fu C, Shah GL. Hospital healthcare resource utilization and costs for chimeric antigen T-cell therapy and autologous hematopoietic cell transplant in patients with large B-cell lymphoma in the United States. Leuk Lymphoma 2024; 65:922-931. [PMID: 38567630 DOI: 10.1080/10428194.2024.2331084] [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: 03/08/2023] [Accepted: 03/11/2024] [Indexed: 04/04/2024]
Abstract
The efficacy of chimeric antigen receptor (CAR) T-cell therapy for large B-cell lymphoma (LBCL) is well-established. This study, using the Premier PINC AI Healthcare Database, assessed hospital costs and healthcare resource utilization (HRU) between CAR T-cell therapy and autologous hematopoietic cell transplant (AHCT) for 733 LBCL patients from 01/01/2017-04/30/2021 (166 CAR T and 567 AHCT from 37 US hospital systems. CAR T-cell therapy had higher index costs but lower non-pharmacy costs, shorter hospital stays, lower ICU utilization than AHCT. The CAR T-cell cohort also presented fewer preparatory costs and HRU. At a 180-day follow-up, AHCT had lower hospitalization rates and costs. Overall, despite higher index costs, CAR T-cell therapy has lower non-pharmacy costs and HRU during the index procedure and requires less preparation time with lower preparation HRUs and costs than AHCT. This has important implications for resource management and informed decision-making for stakeholders.
Collapse
Affiliation(s)
- Chendi Cui
- PINC AI Applied Sciences, Premier Inc, Charlotte, NC, USA
| | | | - Ning Rosenthal
- PINC AI Applied Sciences, Premier Inc, Charlotte, NC, USA
| | - Sally W Wade
- Wade Outcomes Research and Consulting, Salt Lake City, UT, USA
| | - Laura Curry
- PINC AI Applied Sciences, Premier Inc, Charlotte, NC, USA
| | | | - Gunjan L Shah
- Adult Bone Marrow Transplant Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| |
Collapse
|
29
|
Shumnalieva R, Velikova T, Monov S. Expanding the role of CAR T-cell therapy: From B-cell hematological malignancies to autoimmune rheumatic diseases. Int J Rheum Dis 2024; 27:e15182. [PMID: 38742463 DOI: 10.1111/1756-185x.15182] [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: 10/23/2023] [Revised: 04/04/2024] [Accepted: 04/28/2024] [Indexed: 05/16/2024]
Abstract
Chimeric antigen receptor (CAR) T-cell therapy is a form of immunotherapy where the lymphocytes, mostly T-cells, are redirected to specifically recognize and eliminate a target antigen by coupling them with CARs. The binding of CAR and target cell surface antigens leads to vigorous T cell activation and robust anti-tumor immune responses. Areas of implication of CAR T-cell therapies include mainly hematological malignancies (i.e., advanced B-cell cancers); however, recent studies have proven the unprecedented success of the new immunotherapy also in autoimmune rheumatic diseases. We aim to review the recent advances in CAR T-cell therapies in rheumatology but also to address the limitations of their use in the real clinical practice based on the data on their efficacy and safety.
Collapse
Affiliation(s)
- Russka Shumnalieva
- Department of Rheumatology, Clinic of Rheumatology, Medical University-Sofia, Faculty of Medicine, Sofia, Bulgaria
| | - Tsvetelina Velikova
- Medical Faculty, Sofia University "St. Kliment Ohridski"- Sofia, Sofia, Bulgaria
| | - Simeon Monov
- Department of Rheumatology, Clinic of Rheumatology, Medical University-Sofia, Faculty of Medicine, Sofia, Bulgaria
| |
Collapse
|
30
|
Knoedler L, Dean J, Diatta F, Thompson N, Knoedler S, Rhys R, Sherwani K, Ettl T, Mayer S, Falkner F, Kilian K, Panayi AC, Iske J, Safi AF, Tullius SG, Haykal S, Pomahac B, Kauke-Navarro M. Immune modulation in transplant medicine: a comprehensive review of cell therapy applications and future directions. Front Immunol 2024; 15:1372862. [PMID: 38650942 PMCID: PMC11033354 DOI: 10.3389/fimmu.2024.1372862] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2024] [Accepted: 03/22/2024] [Indexed: 04/25/2024] Open
Abstract
Balancing the immune response after solid organ transplantation (SOT) and vascularized composite allotransplantation (VCA) remains an ongoing clinical challenge. While immunosuppressants can effectively reduce acute rejection rates following transplant surgery, some patients still experience recurrent acute rejection episodes, which in turn may progress to chronic rejection. Furthermore, these immunosuppressive regimens are associated with an increased risk of malignancies and metabolic disorders. Despite significant advancements in the field, these IS related side effects persist as clinical hurdles, emphasizing the need for innovative therapeutic strategies to improve transplant survival and longevity. Cellular therapy, a novel therapeutic approach, has emerged as a potential pathway to promote immune tolerance while minimizing systemic side-effects of standard IS regiments. Various cell types, including chimeric antigen receptor T cells (CAR-T), mesenchymal stromal cells (MSCs), regulatory myeloid cells (RMCs) and regulatory T cells (Tregs), offer unique immunomodulatory properties that may help achieve improved outcomes in transplant patients. This review aims to elucidate the role of cellular therapies, particularly MSCs, T cells, Tregs, RMCs, macrophages, and dendritic cells in SOT and VCA. We explore the immunological features of each cell type, their capacity for immune regulation, and the prospective advantages and obstacles linked to their application in transplant patients. An in-depth outline of the current state of the technology may help SOT and VCA providers refine their perioperative treatment strategies while laying the foundation for further trials that investigate cellular therapeutics in transplantation surgery.
Collapse
Affiliation(s)
- Leonard Knoedler
- Department of Plastic, Hand and Reconstructive Surgery, University Hospital Regensburg, Regensburg, Germany
- Division of Plastic Surgery, Department of Surgery, Yale New Haven Hospital, Yale School of Medicine, New Haven, CT, United States
| | - Jillian Dean
- School of Medicine, University of Pittsburgh, Pittsburgh, PA, United States
| | - Fortunay Diatta
- Division of Plastic Surgery, Department of Surgery, Yale New Haven Hospital, Yale School of Medicine, New Haven, CT, United States
| | - Noelle Thompson
- University of Toledo College of Medicine and Life Sciences, Toledo, OH, United States
| | - Samuel Knoedler
- Department of Plastic, Hand and Reconstructive Surgery, University Hospital Regensburg, Regensburg, Germany
| | - Richmond Rhys
- Department of Plastic, Hand and Reconstructive Surgery, University Hospital Regensburg, Regensburg, Germany
| | - Khalil Sherwani
- Department of Hand, Plastic and Reconstructive Surgery, Burn Center, Berufsgenossenschaft (BG) Trauma Center Ludwigshafen, University of Heidelberg, Ludwigshafen, Germany
| | - Tobias Ettl
- Department of Dental, Oral and Maxillofacial Surgery, Regensburg, Germany
| | - Simon Mayer
- University of Toledo College of Medicine and Life Sciences, Toledo, OH, United States
| | - Florian Falkner
- Department of Hand, Plastic and Reconstructive Surgery, Burn Center, Berufsgenossenschaft (BG) Trauma Center Ludwigshafen, University of Heidelberg, Ludwigshafen, Germany
| | - Katja Kilian
- Department of Hand, Plastic and Reconstructive Surgery, Burn Center, Berufsgenossenschaft (BG) Trauma Center Ludwigshafen, University of Heidelberg, Ludwigshafen, Germany
| | - Adriana C. Panayi
- Department of Hand, Plastic and Reconstructive Surgery, Burn Center, Berufsgenossenschaft (BG) Trauma Center Ludwigshafen, University of Heidelberg, Ludwigshafen, Germany
| | - Jasper Iske
- Department of Cardiothoracic and Vascular Surgery, Deutsches Herzzentrum der Charité, Berlin, Germany
- Charité Universitätsmedizin Berlin, Berlin, Germany
| | - Ali-Farid Safi
- Faculty of Medicine, University of Bern, Bern, Switzerland
- Craniologicum, Center for Cranio-Maxillo-Facial Surgery, Bern, Switzerland
| | - Stefan G. Tullius
- Division of Transplant Surgery, Department of Surgery, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA, United States
| | - Siba Haykal
- Department of Plastic, Hand and Reconstructive Surgery, University Hospital Regensburg, Regensburg, Germany
| | - Bohdan Pomahac
- Department of Plastic, Hand and Reconstructive Surgery, University Hospital Regensburg, Regensburg, Germany
| | - Martin Kauke-Navarro
- Department of Plastic, Hand and Reconstructive Surgery, University Hospital Regensburg, Regensburg, Germany
| |
Collapse
|
31
|
Gye A, Lourenco RDA, Goodall S. Discrete Event Simulation to Incorporate Infusion Wait-Time When Assessing Cost-Effectiveness of a Chimeric-Antigen Receptor T Cell Therapy. VALUE IN HEALTH : THE JOURNAL OF THE INTERNATIONAL SOCIETY FOR PHARMACOECONOMICS AND OUTCOMES RESEARCH 2024; 27:415-424. [PMID: 38301961 DOI: 10.1016/j.jval.2024.01.008] [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: 06/28/2023] [Revised: 12/21/2023] [Accepted: 01/11/2024] [Indexed: 02/03/2024]
Abstract
OBJECTIVES The main objective was to use discrete event simulation to model the impact of wait-time, defined as the time between leukapheresis and chimeric antigen receptor (CAR-T) infusion, when assessing the cost-effectiveness of tisagenlecleucel in young patients with relapsed/refractory acute lymphoblastic leukemia. METHODS The movement of patients through the model was determined by parametric time-to-event distributions, with the competing risk of an event determining the costs and quality-adjusted life-years (QALYs) assigned. Cost-effectiveness was expressed using the incremental cost-effectiveness ratio (ICER) for tisagenlecleucel compared with chemotherapy over the lifetime. RESULTS The base case generated a total of 5.79 QALYs and $622 872 for tisagenlecleucel and 1.19 QALYs and $181 219 for blinatumomab, resulting in an ICER of $96 074 per QALY. An increase in mean CAR-T wait-time to 6.20 months reduced the benefit and costs of tisagenlecleucel to 2.78 QALYs and $294 478 because of fewer patients proceeding to infusion, reducing the ICER to $71 112 per QALY. Alternatively, when the cost of tisagenlecleucel was assigned pre-infusion in sensitivity analysis, the ICER increased with increasing wait-time. CONCLUSIONS Under a payment arrangement where CAR-T cost is incurred post-infusion, the loss of benefit to patients is not reflected in the ICER. This may be misguiding to decision makers, where cost-effectiveness ratios are used to guide resource allocation. discrete event simulation is an important tool for economic modeling of CAR-T as it is amenable to capturing the impact of wait-time, facilitating better understanding of factors affecting service delivery and consequently informed decision making to deliver faster access to CAR-T for patients.
Collapse
Affiliation(s)
- Amy Gye
- Centre for Health Economics Research and Evaluation, University of Technology Sydney, Sydney, NSW, Australia.
| | - Richard De Abreu Lourenco
- Centre for Health Economics Research and Evaluation, University of Technology Sydney, Sydney, NSW, Australia
| | - Stephen Goodall
- Centre for Health Economics Research and Evaluation, University of Technology Sydney, Sydney, NSW, Australia
| |
Collapse
|
32
|
Xu S, Wang Q, Ma W. Cytokines and soluble mediators as architects of tumor microenvironment reprogramming in cancer therapy. Cytokine Growth Factor Rev 2024; 76:12-21. [PMID: 38431507 DOI: 10.1016/j.cytogfr.2024.02.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2024] [Accepted: 02/26/2024] [Indexed: 03/05/2024]
Abstract
Navigating the intricate landscape of the tumor microenvironment (TME) unveils a pivotal arena for cancer therapeutics, where cytokines and soluble mediators emerge as double-edged swords in the fight against cancer. This review ventures beyond traditional perspectives, illuminating the nuanced interplay of these elements as both allies and adversaries in cancer dynamics. It critically evaluates the evolving paradigms of TME reprogramming, spotlighting innovative strategies that target the sophisticated network of cytokines and mediators. Special focus is placed on unveiling the therapeutic potential of novel cytokines and mediators, particularly their synergistic interactions with extracellular vesicles, which represent underexplored conduits for therapeutic targeting. Addressing a significant gap in current research, we explore the untapped potential of these biochemical players in orchestrating immune responses, tumor proliferation, and metastasis. The review advocates for a paradigm shift towards exploiting these dynamic interactions within the TME, aiming to transcend conventional treatments and pave the way for a new era of precision oncology. Through a critical synthesis of recent advancements, we highlight the imperative for innovative approaches that harness the full spectrum of cytokine and mediator activities, setting the stage for breakthrough therapies that offer heightened specificity, reduced toxicity, and improved patient outcomes.
Collapse
Affiliation(s)
- Suling Xu
- Department of Dermatology, The First Affiliated Hospital of Ningbo University School of Medicine, Ningbo, Zhejiang 315020, China.
| | - Qingqing Wang
- Institute of Immunology, Zhejiang University School of Medicine, Hangzhou, Zhejiang 310058, China
| | - Wenxue Ma
- Division of Regenerative Medicine, Department of Medicine, Moores Cancer Center, and Sanford Stem Cell Institute, University of California San Diego, La Jolla, CA 92093, USA.
| |
Collapse
|
33
|
Lelarge V, Capelle R, Oger F, Mathieu T, Le Calvé B. Senolytics: from pharmacological inhibitors to immunotherapies, a promising future for patients' treatment. NPJ AGING 2024; 10:12. [PMID: 38321020 PMCID: PMC10847408 DOI: 10.1038/s41514-024-00138-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/17/2023] [Accepted: 01/17/2024] [Indexed: 02/08/2024]
Abstract
The involvement of cellular senescence in the initiation and propagation of diseases is clearly characterized, making the elimination of senescent cells essential to treat age-related diseases. The development of senolytic drugs demonstrated that targeting these cells limits the deterioration of patients' condition, by inducing apoptosis. Nevertheless, the first generations of senolytics which has been developed displayed their activities through specific mechanisms and demonstrated several limitations during clinical development. However, the rational to eliminate senescent cells remains evident, with the necessity to develop specific therapies in a context of diseases and tissues. The evolutions in the field of drug discovery open the way to a new generation of senolytic therapies, such as immunological approaches (CAR-T cells, Antibody-Drug Conjugated or vaccines), which require preliminary steps of research to identify markers specifically expressed on senescent cells, demonstrating promising specific effects. Currently, the preclinical development of these strategies appears more challenging to avoid strong side effects, but the expected results are commensurate with patients' hopes for treatments. In this review, we highlight the fact that the classical senolytic approach based on drug repurposing display limited efficacy and probably reached its limits in term of clinical development. The recent development of more complex therapies and the extension of interest in the domain of senescence in different fields of research allow to extend the possibility to discover powerful therapies. The future of age-related diseases treatment is linked to the development of new approaches based on cell therapy or immunotherapy to offer the best treatment for patients.
Collapse
Affiliation(s)
- V Lelarge
- StarkAge Therapeutics, Campus de l'Institut Pasteur de Lille, 1 rue du Professeur Calmette, 59800, Lille, France
| | - R Capelle
- StarkAge Therapeutics, Campus de l'Institut Pasteur de Lille, 1 rue du Professeur Calmette, 59800, Lille, France
| | - F Oger
- University of Lille, Inserm, CHU Lille, Institut Pasteur de Lille, CNRS, U1283 - UMR 8199 - EGID, 59000, Lille, France
| | - T Mathieu
- StarkAge Therapeutics, Campus de l'Institut Pasteur de Lille, 1 rue du Professeur Calmette, 59800, Lille, France
- Synlab, 60/62 Rue d'Hauteville, 75010, Paris, France
| | - B Le Calvé
- StarkAge Therapeutics, Campus de l'Institut Pasteur de Lille, 1 rue du Professeur Calmette, 59800, Lille, France.
| |
Collapse
|
34
|
Mishra AK, Schmidt TM, Martell EB, Chen AS, Dogru RE, Hematti P, Callander NS. PD1 +TIGIT +2B4 +KLRG1 + Cells Might Underlie T Cell Dysfunction in Patients Treated with BCMA-Directed Chimeric Antigen Receptor T Cell Therapy. Transplant Cell Ther 2024; 30:191-202. [PMID: 37967650 DOI: 10.1016/j.jtct.2023.11.014] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2023] [Revised: 10/07/2023] [Accepted: 11/10/2023] [Indexed: 11/17/2023]
Abstract
Chimeric antigen receptor T cell (CAR-T) therapy has shown rapid, frequent, and deep responses in patients with relapsed/refractory multiple myeloma (RRMM). However, relapse frequently occurs following CAR-T therapy, and the cause of this resistance is not well defined. Among the potential mechanisms of resistance, T cell intrinsic factors may be an important source of failure. Here we used spectral flow cytometry to identify the changes in T cell phenotypes in bone marrow aspirates at different stages of multiple myeloma progression, including cases that relapsed after anti-BCMA CAR-T therapy. We identified completely different T cell phenotypes in RRMM and post CAR-T relapse cases compared to healthy donors and earlier stages of multiple myeloma, novel double-negative CD3+ T cells in RRMM and CAR-T relapsed cases, and differences in CD8 T cell phenotype at the baseline between peripheral blood and bone marrow from healthy donors. We found that the majority of T cells in RRMM patients and significant T cell subsets in post-CAR-T relapsed patients expressed multiple coinhibitory markers, including PD1, TIGIT, 2B4, and KLRG1.
Collapse
Affiliation(s)
- Ameet K Mishra
- Department of Medicine, Division of Hematology and Oncology, School of Medicine and Public Health, University of Wisconsin, Madison, Wisconsin.
| | - Timothy M Schmidt
- Department of Medicine, Division of Hematology and Oncology, School of Medicine and Public Health, University of Wisconsin, Madison, Wisconsin
| | - Ella B Martell
- Department of Medicine, Division of Hematology and Oncology, School of Medicine and Public Health, University of Wisconsin, Madison, Wisconsin
| | - Alex S Chen
- Department of Medicine, Division of Hematology and Oncology, School of Medicine and Public Health, University of Wisconsin, Madison, Wisconsin
| | - Reyna E Dogru
- Department of Medicine, Division of Hematology and Oncology, School of Medicine and Public Health, University of Wisconsin, Madison, Wisconsin
| | - Peiman Hematti
- Department of Medicine, Froedtert Hospital and Medical College of Wisconsin, Milwaukee, Wisconsin
| | - Natalie S Callander
- Department of Medicine, Division of Hematology and Oncology, School of Medicine and Public Health, University of Wisconsin, Madison, Wisconsin
| |
Collapse
|
35
|
Colina AS, Shah V, Shah RK, Kozlik T, Dash RK, Terhune S, Zamora AE. Current advances in experimental and computational approaches to enhance CAR T cell manufacturing protocols and improve clinical efficacy. FRONTIERS IN MOLECULAR MEDICINE 2024; 4:1310002. [PMID: 39086435 PMCID: PMC11285593 DOI: 10.3389/fmmed.2024.1310002] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 10/09/2023] [Accepted: 01/08/2024] [Indexed: 08/02/2024]
Abstract
Since the FDA's approval of chimeric antigen receptor (CAR) T cells in 2017, significant improvements have been made in the design of chimeric antigen receptor constructs and in the manufacturing of CAR T cell therapies resulting in increased in vivo CAR T cell persistence and improved clinical outcome in certain hematological malignancies. Despite the remarkable clinical response seen in some patients, challenges remain in achieving durable long-term tumor-free survival, reducing therapy associated malignancies and toxicities, and expanding on the types of cancers that can be treated with this therapeutic modality. Careful analysis of the biological factors demarcating efficacious from suboptimal CAR T cell responses will be of paramount importance to address these shortcomings. With the ever-expanding toolbox of experimental approaches, single-cell technologies, and computational resources, there is renowned interest in discovering new ways to streamline the development and validation of new CAR T cell products. Better and more accurate prognostic and predictive models can be developed to help guide and inform clinical decision making by incorporating these approaches into translational and clinical workflows. In this review, we provide a brief overview of recent advancements in CAR T cell manufacturing and describe the strategies used to selectively expand specific phenotypic subsets. Additionally, we review experimental approaches to assess CAR T cell functionality and summarize current in silico methods which have the potential to improve CAR T cell manufacturing and predict clinical outcomes.
Collapse
Affiliation(s)
- Alfredo S. Colina
- Department of Microbiology & Immunology, Medical College of Wisconsin, Milwaukee, WI, United States
| | - Viren Shah
- Department of Biomedical Engineering, Medical College of Wisconsin and Marquette University, Milwaukee, WI, United States
| | - Ravi K. Shah
- Department of Medicine, Medical College of Wisconsin, Milwaukee, WI, United States
| | - Tanya Kozlik
- Department of Microbiology & Immunology, Medical College of Wisconsin, Milwaukee, WI, United States
| | - Ranjan K. Dash
- Department of Biomedical Engineering, Medical College of Wisconsin and Marquette University, Milwaukee, WI, United States
| | - Scott Terhune
- Department of Microbiology & Immunology, Medical College of Wisconsin, Milwaukee, WI, United States
- Department of Biomedical Engineering, Medical College of Wisconsin and Marquette University, Milwaukee, WI, United States
| | - Anthony E. Zamora
- Department of Microbiology & Immunology, Medical College of Wisconsin, Milwaukee, WI, United States
- Department of Medicine, Medical College of Wisconsin, Milwaukee, WI, United States
| |
Collapse
|
36
|
Watanabe T. Gene targeted and immune therapies for nodal and gastrointestinal follicular lymphomas. World J Gastroenterol 2023; 29:6179-6197. [PMID: 38186866 PMCID: PMC10768399 DOI: 10.3748/wjg.v29.i48.6179] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/24/2023] [Revised: 11/02/2023] [Accepted: 12/18/2023] [Indexed: 12/27/2023] Open
Abstract
Follicular lymphoma (FL) is the most common indolent B-cell lymphoma (BCL) globally. Recently, its incidence has increased in Europe, the United States, and Asia, with the number of gastrointestinal FL cases expected to increase. Genetic abnormalities related to t(14;18) translocation, BCL2 overexpression, NF-κB pathway-related factors, histone acetylases, and histone methyltransferases cause FL and enhance its proliferation. Meanwhile, microRNAs are commonly used in diagnosing FL and predicting patient prognosis. Many clinical trials on novel therapeutics targeting these genetic abnormalities and immunomodulatory mechanisms have been conducted, resulting in a marked improvement in therapeutic outcomes for FL. Although developing these innovative therapeutic agents targeting specific genetic mutations and immune pathways has provided hope for curative options, FL treatment has become more complex, requiring combinatorial therapeutic regimens. However, optimal treatment combinations have not yet been achieved, highlighting the importance of a complete under-standing regarding the pathogenesis of gastrointestinal FL. Accordingly, this article reviews key research on the molecular pathogenesis of nodal FL and novel therapies targeting the causative genetic mutations. Moreover, the results of clinical trials are summarized, with a particular focus on treating nodal and gastrointestinal FLs.
Collapse
Affiliation(s)
- Takuya Watanabe
- Department of Internal Medicine and Gastroenterology, Watanabe Internal Medicine Aoyama Clinic, Niigata 9502002, Japan
| |
Collapse
|
37
|
McGough SF, Shamas N, Wang J, Jaber M, Swarup B, Blanchet Zumofen MH, Lautié B, Parreira J, Wei MC, Shewade A. Comparative effectiveness between mosunetuzumab monotherapy clinical trial and real-world data in relapsed/refractory follicular lymphoma in third or subsequent lines of systemic therapy. Leuk Lymphoma 2023; 64:2269-2278. [PMID: 37840271 DOI: 10.1080/10428194.2023.2262066] [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: 04/05/2023] [Accepted: 09/14/2023] [Indexed: 10/17/2023]
Abstract
A comparison of clinical outcomes in the third or subsequent line (3 L+) of systemic therapy between a real-world data (RWD) external control cohort and a mosunetuzumab single-arm clinical trial cohort is presented. Data for 3 L + patients with relapsed/refractory follicular lymphoma (FL) were obtained from the mosunetuzumab single-arm trial (n = 90) and a US electronic health records database (n = 158), with patients meeting key eligibility criteria from the trial, balanced on pre-specified prognostic factors. Overall response and complete response rates were 80% and 60% in the mosunetuzumab cohort and 75% and 33% in the RWD cohort, odds ratios of 1.23 (95% CI, 0.52-2.93) and 3.18 (95% CI, 1.41-7.17), respectively. Hazard ratios for progression-free survival and overall survival were 0.82 (95% CI, 0.53-1.27) and 0.43 (95% CI, 0.19-0.94). These findings support a clinically meaningful benefit of mosunetuzumab monotherapy as a chemotherapy-free option for the 3 L + FL population.
Collapse
Affiliation(s)
| | | | - Jue Wang
- Genentech, Inc., South San Francisco, California, USA
| | | | | | | | | | | | - Michael C Wei
- Genentech, Inc., South San Francisco, California, USA
| | | |
Collapse
|
38
|
Vu SH, Pham HH, Pham TTP, Le TT, Vo MC, Jung SH, Lee JJ, Nguyen XH. Adoptive NK Cell Therapy - a Beacon of Hope in Multiple Myeloma Treatment. Front Oncol 2023; 13:1275076. [PMID: 38023191 PMCID: PMC10656693 DOI: 10.3389/fonc.2023.1275076] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2023] [Accepted: 10/18/2023] [Indexed: 12/01/2023] Open
Abstract
Major advances in the treatment of multiple myeloma (MM) have been achieved by effective new agents such as proteasome inhibitors, immunomodulatory drugs, or monoclonal antibodies. Despite significant progress, MM remains still incurable and, recently, cellular immunotherapy has emerged as a promising treatment for relapsed/refractory MM. The emergence of chimeric antigen receptor (CAR) technology has transformed immunotherapy by enhancing the antitumor functions of T cells and natural killer (NK) cells, leading to effective control of hematologic malignancies. Recent advancements in gene delivery to NK cells have paved the way for the clinical application of CAR-NK cell therapy. CAR-NK cell therapy strategies have demonstrated safety, tolerability, and substantial efficacy in treating B cell malignancies in various clinical settings. However, their effectiveness in eliminating MM remains to be established. This review explores multiple approaches to enhance NK cell cytotoxicity, persistence, expansion, and manufacturing processes, and highlights the challenges and opportunities associated with CAR-NK cell therapy against MM. By shedding light on these aspects, this review aims to provide valuable insights into the potential of CAR-NK cell therapy as a promising approach for improving the treatment outcomes of MM patients.
Collapse
Affiliation(s)
- Son Hai Vu
- Hi-Tech Center and Vinmec-VinUni Institute of Immunology, Vinmec Healthcare System, Hanoi, Vietnam
| | - Ha Hong Pham
- Hi-Tech Center and Vinmec-VinUni Institute of Immunology, Vinmec Healthcare System, Hanoi, Vietnam
| | - Thao Thi Phuong Pham
- Hi-Tech Center and Vinmec-VinUni Institute of Immunology, Vinmec Healthcare System, Hanoi, Vietnam
| | - Thanh Thien Le
- Hi-Tech Center and Vinmec-VinUni Institute of Immunology, Vinmec Healthcare System, Hanoi, Vietnam
| | - Manh-Cuong Vo
- Research Center for Cancer Immunotherapy, Chonnam National University Hwasun Hospital, Hwasun, Jeollanamdo, Republic of Korea
| | - Sung-Hoon Jung
- Research Center for Cancer Immunotherapy, Chonnam National University Hwasun Hospital, Hwasun, Jeollanamdo, Republic of Korea
- Department of Hematology-Oncology, Chonnam National University Hwasun Hospital and Chonnam National University Medical School, Hwasun, Jeollanamdo, Republic of Korea
| | - Je-Jung Lee
- Research Center for Cancer Immunotherapy, Chonnam National University Hwasun Hospital, Hwasun, Jeollanamdo, Republic of Korea
- Department of Hematology-Oncology, Chonnam National University Hwasun Hospital and Chonnam National University Medical School, Hwasun, Jeollanamdo, Republic of Korea
| | - Xuan-Hung Nguyen
- Hi-Tech Center and Vinmec-VinUni Institute of Immunology, Vinmec Healthcare System, Hanoi, Vietnam
- College of Health Sciences, VinUniversity, Hanoi, Vietnam
| |
Collapse
|
39
|
Monberg TJ, Borch TH, Svane IM, Donia M. TIL Therapy: Facts and Hopes. Clin Cancer Res 2023; 29:3275-3283. [PMID: 37058256 DOI: 10.1158/1078-0432.ccr-22-2428] [Citation(s) in RCA: 32] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2022] [Revised: 02/20/2023] [Accepted: 03/31/2023] [Indexed: 04/15/2023]
Abstract
After a positive phase III trial, it is evident that treatment with tumor-infiltrating lymphocytes (TIL) is a safe, feasible, and effective treatment modality for patients with metastatic melanoma. Further, the treatment is safe and feasible in diverse solid tumors, regardless of the histologic type. Still, TIL treatment has not obtained the regulatory approvals to be implemented on a larger scale. Therefore, its availability is currently restricted to a few centers worldwide. In this review, we present the current knowledge of TIL therapy and discuss the practical, logistic, and economic challenges associated with implementing TIL therapy on a larger scale. Finally, we suggest strategies to facilitate the widespread implementation of TIL therapy and approaches to develop the next generation of TILs.
Collapse
Affiliation(s)
- Tine J Monberg
- Department of Oncology, National Center for Cancer Immune Therapy (CCIT-DK), Copenhagen University Hospital, Herlev and Gentofte, Herlev, Denmark
| | - Troels H Borch
- Department of Oncology, National Center for Cancer Immune Therapy (CCIT-DK), Copenhagen University Hospital, Herlev and Gentofte, Herlev, Denmark
| | - Inge M Svane
- Department of Oncology, National Center for Cancer Immune Therapy (CCIT-DK), Copenhagen University Hospital, Herlev and Gentofte, Herlev, Denmark
| | - Marco Donia
- Department of Oncology, National Center for Cancer Immune Therapy (CCIT-DK), Copenhagen University Hospital, Herlev and Gentofte, Herlev, Denmark
| |
Collapse
|
40
|
Cappuzzello E, Vigolo E, D’Accardio G, Astori G, Rosato A, Sommaggio R. How can Cytokine-induced killer cells overcome CAR-T cell limits. Front Immunol 2023; 14:1229540. [PMID: 37675107 PMCID: PMC10477668 DOI: 10.3389/fimmu.2023.1229540] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2023] [Accepted: 08/02/2023] [Indexed: 09/08/2023] Open
Abstract
The successful treatment of patients affected by B-cell malignancies with Chimeric Antigen Receptor (CAR)-T cells represented a breakthrough in the field of adoptive cell therapy (ACT). However, CAR-T therapy is not an option for every patient, and several needs remain unmet. In particular, the production of CAR-T cells is expensive, labor-intensive and logistically challenging; additionally, the toxicities deriving from CAR-T cells infusion, such as cytokine release syndrome (CRS) and immune effector cell-associated neurotoxicity syndrome (ICANS), have been documented extensively. Alternative cellular therapy products such as Cytokine-induced killer (CIK) cells have the potential to overcome some of these obstacles. CIK cells are a heterogeneous population of polyclonal CD3+CD56+ T cells with phenotypic and functional properties of NK cells. CIK cell cytotoxicity is exerted in a major histocompatibility complex (MHC)-unrestricted manner through the engagement of natural killer group 2 member D (NKG2D) molecules, against a wide range of hematological and solid tumors without the need for prior antigen exposure or priming. The foremost potential of CIK cells lies in the very limited ability to induce graft-versus-host disease (GvHD) reactions in the allogeneic setting. CIK cells are produced with a simple and extremely efficient expansion protocol, which leads to a massive expansion of effector cells and requires a lower financial commitment compared to CAR-T cells. Indeed, CAR-T manufacturing involves the engineering with expensive GMP-grade viral vectors in centralized manufacturing facilities, whereas CIK cell production is successfully performed in local academic GMP facilities, and CIK cell treatment is now licensed in many countries. Moreover, the toxicities observed for CAR-T cells are not present in CIK cell-treated patients, thus further reducing the costs associated with hospitalization and post-infusion monitoring of patients, and ultimately encouraging the delivery of cell therapies in the outpatient setting. This review aims to give an overview of the limitations of CAR-T cell therapy and outline how the use of CIK cells could overcome such drawbacks thanks to their unique features. We highlight the undeniable advantages of using CIK cells as a therapeutic product, underlying the opportunity for further research on the topic.
Collapse
Affiliation(s)
- Elisa Cappuzzello
- Immunology and Molecular Oncology Unit, Veneto Institute of Oncology IOV - IRCCS, Padova, Italy
| | - Emilia Vigolo
- Immunology and Molecular Oncology Unit, Veneto Institute of Oncology IOV - IRCCS, Padova, Italy
| | - Giulia D’Accardio
- Department of Surgery, Oncology and Gastroenterology, University of Padova, Padova, Italy
| | - Giuseppe Astori
- Advanced Cellular Therapy Laboratory, Department of Hematology, San Bortolo Hospital of Vicenza, Vicenza, Italy
| | - Antonio Rosato
- Immunology and Molecular Oncology Unit, Veneto Institute of Oncology IOV - IRCCS, Padova, Italy
- Department of Surgery, Oncology and Gastroenterology, University of Padova, Padova, Italy
| | - Roberta Sommaggio
- Immunology and Molecular Oncology Unit, Veneto Institute of Oncology IOV - IRCCS, Padova, Italy
- Department of Surgery, Oncology and Gastroenterology, University of Padova, Padova, Italy
| |
Collapse
|
41
|
Watanabe T. Recent advances in treatment of nodal and gastrointestinal follicular lymphoma. World J Gastroenterol 2023; 29:3574-3594. [PMID: 37398889 PMCID: PMC10311612 DOI: 10.3748/wjg.v29.i23.3574] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/13/2023] [Revised: 05/14/2023] [Accepted: 05/22/2023] [Indexed: 06/16/2023] Open
Abstract
Follicular lymphoma (FL) is the most common low-grade lymphoma, and although nodal FL is highly responsive to treatment, the majority of patients relapse repeatedly, and the disease has been incurable with a poor prognosis. However, primary FL of the gastrointestinal tract has been increasingly detected in Japan, especially due to recent advances in small bowel endoscopy and increased opportunities for endoscopic examinations and endoscopic diagnosis. However, many cases are detected at an early stage, and the prognosis is good in many cases. In contrast, in Europe and the United States, gastrointestinal FL has long been considered to be present in 12%-24% of Stage-IV patients, and the number of advanced gastrointestinal cases is expected to increase. This editorial provides an overview of the recent therapeutic advances in nodal FL, including antibody-targeted therapy, bispecific antibody therapy, epigenetic modulation, and chimeric antigen receptor T-cell therapy, and reviews the latest therapeutic manuscripts published in the past year. Based on an understanding of the therapeutic advances in nodal FL, we also discuss future possibilities for gastroenterologists to treat gastrointestinal FL, especially in advanced cases.
Collapse
Affiliation(s)
- Takuya Watanabe
- Department of Internal Medicine and Gastroenterology, Watanabe Internal Medicine Aoyama Clinic, Niigata-city 9502002, Japan
| |
Collapse
|
42
|
Zhang M, Onakpoya I, Rupalla K. International oncology drug approvals for multiregional or single-country clinical trials: A systematic review. Front Med (Lausanne) 2022; 9:1084980. [PMID: 36590932 PMCID: PMC9798114 DOI: 10.3389/fmed.2022.1084980] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2022] [Accepted: 12/01/2022] [Indexed: 12/23/2022] Open
Abstract
Background Cancer remains one of the most common causes of morbidity and mortality worldwide. Multiregional (MRCTs) and single-country clinical trials are two common approaches to support new oncology drug approvals internationally. However, systematic reviews comparing MRCTs with single-country trials for international oncology drug approval are lacking. Methods We searched health agency websites to retrieve all approved oncology drugs from 2010 to 2022. ClinicalTrials.gov was used to retrieve all pivotal study information. We used an adapted version 2 of the Cochrane risk-of-bias tool for randomized trials (RoB 2) and Risk Of Bias In Non-randomized Studies - of Interventions (ROBINS-I) checklist to assess the risk-of-bias in randomized and non-randomized trials, respectively. Results A total of 48 new drugs and biologics (comprising 215 pivotal clinical trials) with initial marketing approval in the United States, European Union, Japan, and China were included. The reporting quality of MRCTs vs. single-country studies was similar. The median time interval for approval was significantly longer for MRCTs than for single-country bridging studies (1,399 vs. 975 days, P < 0.0001), whereas the median time interval for approval was shorter for MRCTs than for single-country standalone studies. The time gap for oncology drugs approved before 2015 was significantly longer than for those approved after 2015. The median timeline for approval in MRCTs involving 3 regions showed the shortest time-to-approval compared with MRCTs involving 4-5 and 1-2 regions. There was no significant difference in the time-to-approval among different tumor types and product types. Conclusion The median time-to-approval of MRCTs was significantly longer than that of single-country bridging studies but shorter than that of single-country standalone studies, primarily involving 3 regions as the most frequent pattern and the shortest time-to-approval to operate MRCTs as a pivotal trial. Single-country bridging studies still provide essential supplements for international oncology drug approvals if MRCTs do not apply. Future studies should explore how to shorten the time-to-approval for MRCTs. Systematic review registration [https://www.researchregistry.com/browsethe-registry#registryofsystematicreviewsmeta-analyses/], identifier [1390].
Collapse
Affiliation(s)
- Min Zhang
- Nuffield Department of Primary Care Health Sciences, University of Oxford, Oxford, United Kingdom,*Correspondence: Min Zhang,
| | - Igho Onakpoya
- Nuffield Department of Primary Care Health Sciences, University of Oxford, Oxford, United Kingdom
| | - Katrin Rupalla
- Ymmunobio AG, Basel, Switzerland,Widler & Schiemann AG, Basel, Switzerland
| |
Collapse
|
43
|
Chacim S, Monjardino T, Cunha JL, Medeiros P, Redondo P, Bento MJ, Mariz JM. Costs, effectiveness, and safety associated with Chimeric Antigen Receptor (CAR) T-cell therapy: Results from a comprehensive cancer center. PLoS One 2022; 17:e0278950. [PMID: 36490296 PMCID: PMC9733886 DOI: 10.1371/journal.pone.0278950] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2022] [Accepted: 11/24/2022] [Indexed: 12/13/2022] Open
Abstract
Real world effectiveness, toxicity and costs analyses from chimeric antigen receptor (CAR)-T cell therapy are of utmost relevance to determine whether and how to offer patients highly personalized immunotherapy. In this study, we aimed at describing CAR T-cells effectiveness, safety and costs in a Portuguese Comprehensive Cancer Center. We performed a retrospective descriptive study of adult patients with relapsed/refractory diffuse large B-cell lymphoma (DLBCL), primary mediastinal B-cell lymphoma and transformed follicular lymphoma referred to CAR T-cell therapy, between May 2019 and February 2021. Rates of treatment response, toxicity and survival (Kaplan-Meier method) were analyzed by intention-to-treat. Direct medical costs stratified by inpatient-care, outpatient-care, and diagnostic-therapeutic procedures (DTP) were derived based on resources used and their respective unit costs. In twenty patients (median age 49.5y; 55%male; 70%DLBCL; 50% with primary refractory disease), best overall and complete response rates were 65.0% and 45.0%, respectively. Median overall (OS) and progression-free survivals were 9.2 and 7.3 months; 12-month OS rate was 42.6% (95%CI:23.2-78.3). Grade≥3 cytokine release syndrome and neurotoxicity occurred in 5.6% and 11.1% of patients, respectively. CAR T-cell therapy expenditure, including adverse events costs, was 7 176 196€, or 286 238€ when excluding drug cost. Median cost for treated patient was 355 165€ with CAR T-cell drug cost accounting for 97.0% of the overall expense. Excluding CAR T-cell acquisition cost, inpatient-care and DTP accounted for 57% and 38% of total cost/patient, respectively. Our findings highlight the heavy economic burden of CAR T-cell therapy driven by drug acquisition costs.
Collapse
Affiliation(s)
- Sérgio Chacim
- Department of Onco-hematology, Portuguese Oncology Institute of Porto (IPO-Porto), Porto, Portugal
- Cancer Biology and Epigenetics Group, Portuguese Oncology Institute of Porto Research Center (CI-IPOP) / RISE@CI-IPOP (Health Research Network) / Porto Comprehensive Cancer Center (Porto.CCC), Porto, Portugal
| | - Teresa Monjardino
- Cancer Epidemiology Group, Portuguese Oncology Institute of Porto Research Center (CI-IPOP) / RISE@CI-IPOP (Health Research Network) / Porto Comprehensive Cancer Center (Porto.CCC), Porto, Portugal
- * E-mail:
| | - José Luís Cunha
- Outcomes Research Lab, Portuguese Oncology Institute of Porto (IPO-Porto), Porto, Portugal
- Management, Outcomes Research, and Economics in Healthcare Group, Portuguese Oncology Institute of Porto Research Center (CI-IPOP) / RISE@CI-IPOP (Health Research Network) / Porto Comprehensive Cancer Center (Porto.CCC), Porto, Portugal
| | - Pedro Medeiros
- Outcomes Research Lab, Portuguese Oncology Institute of Porto (IPO-Porto), Porto, Portugal
- Management, Outcomes Research, and Economics in Healthcare Group, Portuguese Oncology Institute of Porto Research Center (CI-IPOP) / RISE@CI-IPOP (Health Research Network) / Porto Comprehensive Cancer Center (Porto.CCC), Porto, Portugal
- Medicine and Oncological Medicine Departments Management, Portuguese Oncology Institute of Porto (IPO-Porto), Porto, Portugal
| | - Patrícia Redondo
- Outcomes Research Lab, Portuguese Oncology Institute of Porto (IPO-Porto), Porto, Portugal
- Management, Outcomes Research, and Economics in Healthcare Group, Portuguese Oncology Institute of Porto Research Center (CI-IPOP) / RISE@CI-IPOP (Health Research Network) / Porto Comprehensive Cancer Center (Porto.CCC), Porto, Portugal
| | - Maria José Bento
- Cancer Epidemiology Group, Portuguese Oncology Institute of Porto Research Center (CI-IPOP) / RISE@CI-IPOP (Health Research Network) / Porto Comprehensive Cancer Center (Porto.CCC), Porto, Portugal
- Department of Epidemiology, Portuguese Oncology Institute of Porto (IPO-Porto), Porto, Portugal
- Department of Population Studies, ICBAS-School of Medicine and Biomedical Sciences, University of Porto (ICBAS-UP), Porto, Portugal
| | - José Mário Mariz
- Department of Onco-hematology, Portuguese Oncology Institute of Porto (IPO-Porto), Porto, Portugal
| |
Collapse
|
44
|
Banerjee R, Lee SS, Cowan AJ. Innovation in BCMA CAR-T therapy: Building beyond the Model T. Front Oncol 2022; 12:1070353. [PMID: 36505779 PMCID: PMC9729952 DOI: 10.3389/fonc.2022.1070353] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2022] [Accepted: 11/08/2022] [Indexed: 11/25/2022] Open
Abstract
Autologous chimeric antigen receptor T-cell (CAR-T) therapies targeting B-cell maturation antigen (BCMA) have revolutionized the field of multiple myeloma in the same way that the Ford Model T revolutionized the original CAR world a century ago. However, we are only beginning to understand how to improve the efficacy and usability of these cellular therapies. In this review, we explore three automotive analogies for innovation with BCMA CAR-T therapies: stronger engines, better mileage, and hassle-free delivery. Firstly, we can build stronger engines in terms of BCMA targeting: improved antigen binding, tools to modulate antigen density, and armoring to better reach the antigen itself. Secondly, we can improve "mileage" in terms of response durability through ex vivo CAR design and in vivo immune manipulation. Thirdly, we can implement hassle-free delivery through rapid manufacturing protocols and off-the-shelf products. Just as the Model T set a benchmark for car manufacturing over 100 years ago, idecabtagene vicleucel and ciltacabtagene autoleucel have now set the starting point for BCMA CAR-T therapy with their approvals. As with any emerging technology, whether automotive or cellular, the best in innovation and optimization is yet to come.
Collapse
Affiliation(s)
- Rahul Banerjee
- Division of Medical Oncology, Department of Medicine, University of Washington, Seattle, WA, United States
- Clinical Research Division, Fred Hutchinson Cancer Center, Seattle, WA, United States
| | - Sarah S. Lee
- Division of Medical Oncology, Department of Medicine, University of Washington, Seattle, WA, United States
- Clinical Research Division, Fred Hutchinson Cancer Center, Seattle, WA, United States
| | - Andrew J. Cowan
- Division of Medical Oncology, Department of Medicine, University of Washington, Seattle, WA, United States
- Clinical Research Division, Fred Hutchinson Cancer Center, Seattle, WA, United States
| |
Collapse
|
45
|
Lyou Y, Dorff TB. Chimeric Antigen Receptor (CAR) T-cell Treatment in Renal Cell Carcinoma: Current clinical trials and future directions. KIDNEY CANCER 2022. [DOI: 10.3233/kca-220001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
Renal cell carcinoma (RCC) has long been found to be responsive to immunotherapy. While high dose interleukin-2 resulted in some durable remissions, this treatment has largely been replaced by immune checkpoint inhibitor therapy, due to the safer toxicity profile and emerging evidence for long term remissions. However, the majority of patients continue to face disease progression and death from metastatic RCC. Chimeric antigen receptor T-cells (CAR T) represent the next step in immunotherapy for this malignancy and hold promise for a higher rate of durable remissions. The realization of this therapeutic strategy for RCC will require identification of the best tumor antigen and T cell modifications and will depend on achieving remissions with an acceptable toxicity profile. This review summarizes current CAR T-cell treatment targets and clinical trials for metastatic RCC, highlighting the potential therapeutic impact as well as obstacles to successful development.
Collapse
Affiliation(s)
- Yung Lyou
- Department of Hematology-Oncology, Providence St. Jude Crosson Cancer Institute, Fullerton, CA, USA
| | - Tanya B. Dorff
- Department of Medical Oncology and Experimental Therapeutics, City of Hope Comprehensive Cancer Center, Duarte, CA, USA
| |
Collapse
|
46
|
Horgan D, Walewski J, Aurer I, Visco C, Giné E, Fetica B, Jerkeman M, Kozaric M, da Silva MG, Dreyling M. Tackling Mantle Cell Lymphoma in Europe. Healthcare (Basel) 2022; 10:1682. [PMID: 36141294 PMCID: PMC9498856 DOI: 10.3390/healthcare10091682] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2022] [Revised: 08/24/2022] [Accepted: 08/31/2022] [Indexed: 11/17/2022] Open
Abstract
An expert panel convened by the European Alliance for Personalized Medicine (EAPM) reflected on achievements and outstanding challenges in Europe in mantle cell lymphoma (MCL). Through the prism of member state experience, the panel noted advances in outcomes over the last decade, but highlighted issues constituting barriers to better care. The list notably included availability of newer treatments, infrastructure and funding for related testing, and shortages of relevant skills and of research support. The prospect of improvements was held to reside in closer coordination and cooperation within and between individual countries, and in changes in policy and scale of investment at both national and EU levels.
Collapse
Affiliation(s)
- Denis Horgan
- European Alliance for Personalised Medicine, 1040 Brussels, Belgium
| | - Jan Walewski
- The Maria Sklodowska-Curie National Research Institute of Oncology, 00-001 Warszawa, Poland
| | - Igor Aurer
- Division of Hematology, Department of Internal Medicine, University Hospital Center Zagreb, 10000 Zagreb, Croatia
| | - Carlo Visco
- Department of Medicine, Section of Hematology, University of Verona, 37129 Verona, Italy
| | - Eva Giné
- Instituto Clínic de Enfermedades Hematológicas y Oncológicas, Hospital Clínic de Barcelona, 08036 Barcelona, Spain
| | - Bogdan Fetica
- Department of Pathology, Institute of Oncology “Prof. Dr. Ion Chiricuta” Cluj-Napoca, 400015 Cluj-Napoca, Romania
| | - Mats Jerkeman
- Department of Oncology, Institute of Clinical Sciences, Lund University and Skane, University Hospital, BMC F12, 221 84 Lund, Sweden
| | - Marta Kozaric
- European Alliance for Personalised Medicine, 1040 Brussels, Belgium
| | - Maria Gomes da Silva
- Haematology Unit, Instituto Portugues de Oncologia de Lisboa Francisco Gentil, 1099-023 Lisbon, Portugal
| | - Martin Dreyling
- Medical Clinic III, Groβhadern Clinic, Ludwig-Maximilians-Universität, 81377 Munich, Germany
| |
Collapse
|