RESUMO
Aim: Cost-effectiveness analysis (CEA) was performed to compare axicabtagene ciloleucel (axi-cel) with tisagenlecleucel (tisa-cel) and lisocabtagene (liso-cel) for treatment of relapsed or refractory large B-cell lymphoma in adult patients after ≥2 lines of therapy in Japan. Materials & methods: Cost-effectiveness analysis was conducted using the partition survival mixture cure model based on the ZUMA-1 trial and adjusted to the JULIET and TRANSCEND trials using matching-adjusted indirect comparisons. Results & conclusion: Axi-cel was associated with greater incremental life years (3.13 and 2.85) and incremental quality-adjusted life-years (2.65 and 2.24), thus generated lower incremental direct medical costs (-$976.29 [-¥137,657] and -$242.00 [-¥34,122]), compared with tisa-cel and liso-cel. Axi-cel was cost-effective option compared with tisa-cel and liso-cel from a Japanese payer's perspective.
[Box: see text].
Assuntos
Análise Custo-Benefício , Anos de Vida Ajustados por Qualidade de Vida , Humanos , Japão/epidemiologia , Masculino , Feminino , Linfoma Difuso de Grandes Células B/tratamento farmacológico , Linfoma Difuso de Grandes Células B/economia , Linfoma Difuso de Grandes Células B/mortalidade , Antígenos CD19/economia , Antígenos CD19/imunologia , Antígenos CD19/uso terapêutico , Receptores de Antígenos de Linfócitos T/uso terapêutico , Imunoterapia Adotiva/economia , Imunoterapia Adotiva/métodos , Pessoa de Meia-Idade , Adulto , Vacinas Anticâncer/economia , Vacinas Anticâncer/administração & dosagem , Idoso , Produtos Biológicos/economia , Produtos Biológicos/uso terapêutico , Análise de Custo-EfetividadeRESUMO
BACKGROUND: With the recent Food & Drug Administration (FDA) approval of cellular therapy that requires product manipulation prior to administration in combination with a short stability window, the need was identified for local dose preparation within the pharmacy rather than the off-site stem cell processing laboratory. This approval gave rise to assessment of regulatory standards surrounding cellular therapy, evaluation and revision of current standard operating procedures and policies with formal process validation, assessment of occupational exposure mitigation and safety considerations, and development of staff training and education. OBJECTIVE: To describe and provide insight into the stepwise process of FACT validation and onboarding of commercially available cellular therapy products that require sterile compounding manipulation within a pharmacy prior to administration. DISCUSSION: A multidisciplinary effort is required to attain FACT certification and implement pharmacist compounding of cellular therapy products.1 Local preparation within a pharmacy facilitates a sound operational workflow and provides a pathway to perform aseptic manipulations of cellular therapy products safely and efficiently. CONCLUSION: Safe and successful administration of cellular therapies handled and compounded by pharmacy department staff along with program validation requires a preemptive review utilizing a multidisciplinary approach for process development. This manuscript will provide a foundation based on consistency and transparency in effective cellular therapy sterile compounding and aseptic manipulation, proper handling and disposal procedures, increased communication through creation and optimization of treatment plans and order-sets, standardized medical center staff education, and development of policies and standard operating procedures for the entire health care team.
RESUMO
Chimeric Antigen Receptor (CAR)-modified T lymphocytes represent one of the most innovative and promising approaches to treating hematologic malignancies. CAR-T cell therapy is currently being used for the treatment of relapsed/refractory (r/r) B-cell malignancies including Acute Lymphoblastic Leukemia, Large B-Cell Lymphoma, Follicular Lymphoma, Multiple Myeloma and Mantle Cell Lymphoma. Despite the unprecedented clinical success, one of the major issues of the approved CAR-T cell therapy - tisagenlecleucel, axicabtagene, lisocabtagene, idecabtagene, ciltacabtagene and brexucabtagene - is the uncertainty about its persistence which in turn could lead to weak or no response to therapy with malignancy recurrence. Here we show that the prognosis of patients who do not respond to CAR-T cell therapy is still an unmet medical need. We performed a systematic review and meta-analysis collecting individual data on Duration of Response from at least 12-month follow-up studies. We found that the pooled prevalence of relapse within the first 12 months after CAR-T infusion was 61% (95% CI, 43%-78%); moreover, one year after the infusion, the analysis highlighted a pooled prevalence of relapse of 24% (95% CI, 11%-42%). Our results suggest that identifying potential predictive biomarkers of response to CAR-T therapy, especially for patients affected by the advanced stage of blood malignancies, could lead to stratification of the eligible population to that therapy, recognizing which patients will benefit and which will not, helping regulators to make decision in that way.
Assuntos
Neoplasias Hematológicas , Mieloma Múltiplo , Receptores de Antígenos Quiméricos , Humanos , Adulto , Linfócitos T , Neoplasias Hematológicas/terapia , Doença Crônica , Recidiva , Terapia Baseada em Transplante de Células e TecidosRESUMO
Lisocabtagene maraleucel (liso-cel) is one of the three US FDA-approved chimeric antigen receptor T-cell therapies for the treatment of relapsed/refractory (R/R) large B-cell lymphoma (LBCL). TRANSCEND is the landmark trial that led to the approval of liso-cel in the third-line setting for R/R diffuse LBCL, primary mediastinal B-cell lymphoma, follicular lymphoma grade 3B and transformed lymphoma. The TRANSFORM and PILOT studies evaluated the use of liso-cel in the second-line treatment of R/R LBCL. This review details the structure and manufacturing process of liso-cel that make it distinct from other approved chimeric antigen receptor constructs, outlines results from landmark trials of liso-cel in LBCL and discusses liso-cel toxicity.
Chimeric antigen receptor (CAR) T-cell therapy is a type of treatment for large B-cell lymphoma (LBCL). CAR T involves modifying a patient's immune cells (T cells, specifically), so that they can attack cancer cells and destroy them. Lisocabtagene maraleucel (liso-cel) is a type of CAR T-cell therapy that has been approved to treat patients with LBCL who have already received at least one line of treatment for their lymphoma. This article outlines the structure and manufacturing process of liso-cel and discusses how it differs from other approved CAR T-cell therapies. It also summarizes the clinical data for liso-cel in the treatment of LBCL, as well as its safety and expected side effects.
Assuntos
Linfoma Folicular , Linfoma Difuso de Grandes Células B , Linfoma não Hodgkin , Receptores de Antígenos Quiméricos , Humanos , Linfoma Difuso de Grandes Células B/tratamento farmacológico , Linfócitos B , Imunoterapia Adotiva/efeitos adversos , Antígenos CD19RESUMO
Patients with refractory/early relapsed aggressive lymphomas belong to the most difficult-to-treat patients with hematological neoplasia. The prognosis of such patients is poor and novel treatment approches are urgently needed. Autologous chimeric antigen receptor T-cell therapy is a promising new therapy option that is approved after the failure of two lines of chemotherapy. Recently, the results of three different CART studies (ZUMA-7, TRANSFORM, and BELINDA) were published. Two of them were positive and one was negative, which created a mix of disappointment, confusion, and irritation. In this article, we are analyzing the data of all three trials and shed light on the differences between the studies which may facilitate an easier understanding of the results and relevance of CART in aggressive large B-cell lymphoma.
Assuntos
Antígenos CD19 , Linfoma Difuso de Grandes Células B , Humanos , Antígenos CD19/efeitos adversos , Imunoterapia Adotiva/efeitos adversos , Linfoma Difuso de Grandes Células B/tratamento farmacológicoRESUMO
BACKGROUND AIMS: Autologous chimeric antigen receptor (CAR) T-cell therapies have demonstrated substantial clinical benefit across several hematologic malignancies. However, patient-to-patient variability and heterogeneity of starting cellular material across patient populations and disease indications pose challenges to manufacturing consistency. Lisocabtagene maraleucel (liso-cel) is an autologous, CD19-directed, defined-composition, 4-1BB CAR T-cell product administered at equal target doses of CD8+ and CD4+ CAR+ T cells. Here the authors describe the optimization of the liso-cel manufacturing platform for product quality and consistency. METHODS: Leukapheresis starting materials were collected from patients with large B-cell lymphoma, mantle cell lymphoma or chronic lymphocytic leukemia treated with liso-cel in clinical trials (NCT02631044 and NCT03331198). The liso-cel manufacturing process involves selection of CD8+ and CD4+ T cells from leukapheresis material followed by independent CD8+ and CD4+ T-cell activation, transduction, expansion, formulation and cryopreservation. Multivariate design of experimental approaches was utilized to optimize process conditions at both specific unit operations and across the process. Flow cytometry methods were used to assess cellular composition, memory phenotypes and cell proliferation. Antigen-specific functions, including cytokine secretion, cytolytic activity and proliferation, were assessed using endpoint assays after independent stimulation of CD8+ and CD4+ CAR+ T-cell product components. RESULTS: Reductions in process duration time, optimization of drug product container and formulation and activation signal optimization led to significantly increased CAR+ T-cell product viability. The heterogeneity of patient-derived starting material, including low absolute lymphocyte counts in some samples, was reduced through early T-cell purification, leading to median T-cell frequencies >95% in selected materials across disease indications and limited non-T-cell impurities. These changes further increased lineage purity in CD8+ and CD4+ CAR+ T-cell drug products. CD8+ and CD4+ CAR+ T-cell component lot functional profiles demonstrated multifunctional mechanisms of action, including differential cytokine release, differential cytolytic kinetics and high frequencies of proliferating cells. Correlative analyses demonstrated strong underlying associations between starting material attributes and final CAR+ T-cell product phenotype. CONCLUSIONS: Despite substantial heterogeneity of starting leukapheresis material quality/composition between individual patients and across disease indications/histologies, the liso-cel manufacturing platform is robust and capable of generating a consistent drug product from diverse starting materials with a single manufacturing platform.
Assuntos
Receptores de Antígenos Quiméricos , Antígenos CD19 , Linfócitos T CD8-Positivos , Ensaios Clínicos como Assunto , Citocinas , Humanos , Imunoterapia Adotiva/métodos , Receptores de Antígenos de Linfócitos TRESUMO
Progressive multifocal leukoencephalopathy (PML) is a life-threatening infection of the central nervous system in immunocompromised patients, with an established predilection in non-Hodgkin's lymphoma and stem cell transplant recipients. In the era of chimeric antigen receptor T-cell therapy (CAR T-cell), the occurrence of new-onset neurological symptoms and encephalopathy in this patient population can be attributed to a variety of factors, including therapy-related neurotoxicity or disease progression. PML has not been implicated as a common cause of encephalopathy in CAR T-cell therapy recipients, and the identification of such rare infections is important to guide prognosis and treatment decisions. We hereby report the first case of late occurrence of PML, over one year after CAR T-cell therapy, for a patient with relapsed large B-cell lymphoma.
Assuntos
Imunoterapia Adotiva/efeitos adversos , Leucoencefalopatia Multifocal Progressiva/diagnóstico , Leucoencefalopatia Multifocal Progressiva/etiologia , Antígenos CD19/imunologia , Encéfalo/diagnóstico por imagem , Encéfalo/patologia , Feminino , Humanos , Imunoterapia Adotiva/métodos , Pessoa de Meia-Idade , Neuroimagem , Receptores de Antígenos Quiméricos/imunologiaRESUMO
In 1891, Dr. William B. Coley, an American surgeon, made a compelling observation that immune system can be triggered to shrink tumors. The quest to exploit the power of immunotherapy however was forestalled by an era of chemotherapy that ensued. During World War II, the accidental sinking of a US naval ship led to a group of sailors developing pancytopenia due to poisoning from mustard gas (nitrogen mustard). The observation prompted wide-scale screening of these chemical compounds with cytotoxic potential; further clinical trials led to the first Food and Drug Administration (FDA) approval of a chemotherapy drug, nitrogen mustard. Immunotherapy field took further impetus, not until the last two decades, due to our deeper understanding of the immune system and the cellular and molecular pathways leading to tumor development. Two groundbreaking therapies which have shown great promise in this field involve "taking the breaks off" and "pushing the pedal" of the immune system. These therapies, namely, immune checkpoint inhibitors and adoptive cell therapy, respectively, have been successful in a variety of malignancies, while the former mostly in solid tumors and the latter in hematological malignancies.
Assuntos
Imunoterapia Adotiva , Neoplasias , Humanos , Neoplasias/tratamento farmacológico , Linfócitos TRESUMO
CAR-T (chimeric antigens receptor-T) cell therapy is a breakthrough therapy of the twenty-first century for the management of different malignancies including lymphomas and leukemias. Numeral trials are underway to understand the optimal CAR-T cell design and dose to maximize efficacy and mitigate toxicity. Currently two CAR-T cell therapy products, axicabtagene ciloleucel and tisagenlecleucel, are approved by the US Food and Drug Administration, which have shown excellent responses in otherwise poor prognostic lymphomas and leukemias. The favorable outcomes achieved of this therapy were noted to be durable during long-term follow-up. Understanding the challenges associated with manufacturing and the reasons for T cell failure including poor T cell expansion, persistence, and tumor resistance are critical for its wide-scale application in order to attain the full potential of this novel therapy. Here we review the salient features of the different CAR-T products and discuss the pivotal trials that led to its approval.
Assuntos
Imunoterapia Adotiva , Neoplasias/imunologia , Neoplasias/terapia , Receptores de Antígenos Quiméricos , Linfócitos T , Humanos , Leucemia/imunologia , Leucemia/terapia , Linfoma/imunologia , Linfoma/terapiaRESUMO
Approximately 60-70% of patients with large B cell lymphoma (LBCL) achieve long-term remission or a cure after initial treatment. However, patients who relapse or are refractory to initial treatment have a poor prognosis. Chimeric antigen receptor (CAR) T cell therapy has recently attracted attention for its potential to provide a cure or long-term remission even for LBCL that has relapsed or is refractory to conventional chemotherapy. Currently, three CAR T cell products are clinically available for LBCL: tisagenlecleucel (tisa-cel), axicabtagene ciloleucel (axi-cel) and lisocabtagene maraleucel (liso-cel). These CAR T cell products were initially approved as third- or later-line therapies worldwide. Recently, axi-cel and liso-cel have become feasible as second-line therapies for patients with early relapsed or refractory disease after first-line chemotherapy. Although a large body of data on CAR T cell therapy has been accumulated, the clinical question of how to choose between these three available CAR T cell products has yet to be resolved. The appropriate approach to treatment selection for patients who relapse after CAR T cell therapy also remains unclear. This review discusses treatment strategies to maximize the benefits of CAR T cell therapy.
Assuntos
Imunoterapia Adotiva , Linfoma Difuso de Grandes Células B , Humanos , Imunoterapia Adotiva/métodos , Linfoma Difuso de Grandes Células B/terapia , Linfoma Difuso de Grandes Células B/imunologia , Receptores de Antígenos Quiméricos/imunologia , Receptores de Antígenos de Linfócitos T , Produtos Biológicos/uso terapêuticoRESUMO
Follicular lymphoma (FL) is the most common indolent non-Hodgkin lymphoma. Significant unmet need remains for patients with relapsed/refractory FL after ≥3 lines of prior therapy. While recent advancements have likely improved the survival of patients with FL, most patients will eventually relapse. The treatment of patients with FL after multiple relapses or those with refractory disease has historically led to lower overall response rates (ORR) and shorter progression-free survival (PFS) with each subsequent line of therapy. New treatments with high ORR and durable PFS are needed in this setting, particularly in patients that progress within 2 years of first line chemoimmunotherapy (POD24) and/or those refractory chemoimmunotherapy. Chimeric antigen receptor T-cell therapies targeting the B-cell antigen CD-19 have shown to be an efficacious treatment option for both heavily pretreated patients and/or patients with refractory FL, resulting in a high ORR and durable remissions.
RESUMO
The safety and efficacy of CAR T-cell therapy are unknown in pediatric and adolescent patients with relapsed or refractory primary mediastinal large B-cell lymphoma (R/R PMBCL) which is associated with dismal prognosis. Here, we present a case report of a 16-year-old patient with R/R PMBCL treated with lisocabtagene maraleucel including correlative studies. Patient achieved complete response at 6 months without cytokine release syndrome and immune effector cell-associated neurotoxicity syndrome. She only experienced mild cytopenias, requiring filgrastim once. This report highlights the safety and efficacy of lisocabtagene maraleucel in this population, warranting prospective studies to improve clinical outcomes.
RESUMO
Historically, salvage chemoimmunotherapy with consolidative autologous hematopoietic stem cell transplantation (ASCT) was the only potentially curative therapeutic option for patients with relapsed/refractory large B-cell lymphoma (LBCL). Treatment options were few and outcomes poor for patients whose lymphoma failed to respond to salvage chemotherapy/ASCT and for patients not eligible for ASCT. The approval of chimeric antigen receptor (CAR) T-cell therapy for relapsed/refractory LBCL revolutionized the treatment landscape with unprecedented response rates and durability of responses. As a result, earlier intervention with CAR T-cell therapy has been explored, and the enthusiasm for CAR T-cell therapy has overshadowed ASCT. In this article, we will review the data available for ASCT and CAR T-cell therapy in relapsed LBCL and will examine the role for ASCT in relapsed/refractory LBCL in the era of CAR T-cell therapy.
RESUMO
Chimeric antigen receptor (CAR) T-cell therapy has become a powerful treatment option in B-cell and plasma cell malignancies, and many patients have benefited from its use. To date, six CAR T-cell products have been approved by the FDA and EMA, and many more are being developed and investigated in clinical trials. The whole field of adoptive cell transfer has experienced an unbelievable development process, and we are now at the edge of a new era of immune therapies that will have its impact beyond hematologic malignancies. Areas of interest are, e.g., solid oncology, autoimmune diseases, infectious diseases, and others. Although much has been achieved so far, there is still a huge effort needed to overcome significant challenges and difficulties. We are witnessing a rapid expansion of knowledge, induced by new biomedical technologies and CAR designs. The era of CAR T-cell therapy has just begun, and new products will widen the therapeutic landscape in the future. This review provides a comprehensive overview of the clinical applications of CAR T-cells, focusing on the approved products and emphasizing their benefits but also indicating limitations and challenges.
RESUMO
Bendamustine has been retrospectively shown to be an effective and safe lymphodepletion regimen prior to the anti-CD19 chimeric antigen receptor T cell (CART) products tisagenlecleucel and axicabtagene ciloleucel, as well as the anti-BCMA CART products idecabtagene vicleucel and ciltacabtagene autoleucel. However, bendamustine as lymphodepletion prior to lisocabtagene maraleucel (liso-cel), a 4-1BB co-stimulated, fixed CD4:CD8 ratio anti-CD19 CART product, has not been described yet. Thus, we studied a cohort of sequentially-treated patients with large B-cell lymphomas who received bendamustine lymphodepletion before liso-cel at the University of Pennsylvania between 5/2021 and 12/2023 (n = 31). Patients were evaluated for toxicities and responses. Of note, 7 patients (22.6%) would have dnot met the inclusion criteria for the registrational liso-cel clinical trials, mostly due to older age. Overall and complete response rates were 76.9% and 73.1%, respectively. At a median follow-up of 6.3 months, the 6-month progression-free and overall survival were 59.9% and 91.1%, respectively. Rates of cytokine-release syndrome (CRS) and neurotoxicity (ICANS) of any grade were 9.7% and 9.7%, respectively, with no grade ≥ 3 events. No infections were reported during the first 30 days following liso-cel infusion. Neutropenia ≥ grade 3 was observed in 29.0% of patients; thrombocytopenia ≥ grade 3 occurred in 9.7%. In conclusion, bendamustine lymphodepletion before liso-cel appears to be a strategy that can drive tumor responses while ensuring a mild toxicity profile.
Assuntos
Cloridrato de Bendamustina , Imunoterapia Adotiva , Humanos , Cloridrato de Bendamustina/uso terapêutico , Pessoa de Meia-Idade , Masculino , Feminino , Idoso , Imunoterapia Adotiva/métodos , Imunoterapia Adotiva/efeitos adversos , Estudos Retrospectivos , Adulto , Linfoma Difuso de Grandes Células B/tratamento farmacológico , Antineoplásicos Alquilantes/uso terapêutico , Antineoplásicos Alquilantes/efeitos adversos , Produtos Biológicos/uso terapêutico , Produtos Biológicos/efeitos adversos , Idoso de 80 Anos ou mais , Resultado do TratamentoRESUMO
INTRODUCTION: Chimeric antigen receptor T (CAR T) cell therapy epitomizes the success of T cell engineering. Today, it is an integral component of the treatment algorithm for various types of B-cell non-Hodgkin lymphoma (NHL). Large B-cell lymphoma (LBCL) is the most common subtype of NHL accounting for 30-35% of cases. A lack of response to second-line therapy portends a poor prognosis as only 7-15% of patients attain complete remission (CR) with subsequent conventional chemoimmunotherapy. AREAS COVERED: Lisocabtagene maraleucel (liso-cel) is an autologous CD-19 directed CAR T-cell product with a 4-1BB co-stimulatory domain administered as a sequential infusion of 2 separately manufactured components: CD8+ and CD4+ CAR T-cells in equal doses. Liso-cel showed an impressive objective response rate of 73% (CR = 53%) in patients who had received a median of 3 prior therapies. Median time-to-first CR or partial response (PR) was 1 month. EXPERT OPINION: When evaluated in the second line setting in LBCL, liso-cel demonstrated superior event-free survival (EFS) versus standard of care. While acknowledging that choice of a particular CAR T-cell is based chiefly on familiarity of the treating physician with a specific product, liso-cel definitely represents an important addition to the treatment armamentarium of R/R LBCL whether in the second-line setting or beyond.
Assuntos
Linfoma Difuso de Grandes Células B , Receptores de Antígenos Quiméricos , Humanos , Linfoma Difuso de Grandes Células B/terapia , Imunoterapia Adotiva , Linfócitos T , ImunoterapiaRESUMO
INTRODUCTION: The objective of this study was to evaluate the cost-effectiveness of lisocabtagene maraleucel (liso-cel) versus other available chimeric antigen receptor T-cell therapies, including axicabtagene ciloleucel (axi-cel) and tisagenlecleucel (tisa-cel), in patients who had received at least two prior therapies from a United States (US) commercial third-party payer perspective. METHODS: To capture this heterogeneity in survival outcomes, we used mixture cure models to extrapolate progression-free survival (PFS) and overall survival (OS). Patient-level data from TRANSCEND NHL 001 for liso-cel and reconstructed patient-level data from ZUMA-1 for axi-cel, JULIET for tisa-cel, and SCHOLAR-1 for salvage chemotherapy, derived using the Guyot method, were used for OS and PFS. The model included adverse events associated with liso-cel, axi-cel, and tisa-cel. RESULTS: Liso-cel was less costly (incremental cost of - $74,980) and marginally more effective (0.002 incremental quality-adjusted life-years [QALY]) than axi-cel and had an incremental cost of $67,925 and 2.02 incremental QALYs over tisa-cel in the base case. Results remained consistent in sensitivity analyses, with the liso-cel OS cure fraction being the main driver of cost-effectiveness compared with both axi-cel and tisa-cel. CONCLUSION: This analysis estimated that liso-cel is cost-effective compared with tisa-cel and axi-cel from a commercial US payer perspective.
Assuntos
Linfoma Difuso de Grandes Células B , Humanos , Análise Custo-Benefício , Imunoterapia AdotivaRESUMO
Chimeric antigen receptor (CAR) T-cell therapy is currently approved for the treatment of B-cell non-Hodgkin lymphomas and B-cell acute lymphoblastic leukemia. Prolonged hematological toxicity is an emergent concern following CAR T cells and occurred in 30% of patients with unknown mechanism. Few cases of myelodysplastic syndrome (MDS) following CAR T-cell therapy were reported and attributed to previous chemotherapies in heavily pretreated patients. The authors report the case of a patient with diffuse large B-cell lymphoma treated with axicabtagene ciloleucel who developed prolonged hematological toxicity by day 28. During the follow-up, the diagnosis of MDS was made. The patient underwent allogenic hematological stem cell transplantation. The patient remains in complete remission of his lymphoma and MDS 19 months after hematological stem cell transplantation.
Chimeric antigen receptor (CAR) T cell is a new type of immunotherapy that was recently validated for the treatment of some types of B-cell lymphoma and leukemia. One of the most recently reported side effects of CAR T cells is the appearance of anemia, thrombocytopenia and/or neutropenia lasting for a long duration. The authors report the case of a patient treated with CAR T cells for non-Hodgkin lymphoma who developed prolonged hematological toxicity. During follow-up, the diagnosis of myelodysplastic syndrome was made and the patient underwent allogenic bone marrow transplantation and remains in complete remission at last follow-up.
Assuntos
Transplante de Células-Tronco Hematopoéticas , Linfoma Difuso de Grandes Células B , Síndromes Mielodisplásicas , Receptores de Antígenos Quiméricos , Humanos , Imunoterapia Adotiva/efeitos adversos , Antígenos CD19/uso terapêutico , Receptores de Antígenos de Linfócitos TRESUMO
INTRODUCTION: The new class of chimeric antigen receptor T-cell has emboldened health-care professionals and patients for a more effective treatment of hematological malignancies, indicatively lymphoma, acute lymphoblastic leukemia, and myeloma. Nevertheless, their burgeoning procurement costs comprise a litmus stress for health systems across the globe. In this context, this systematic review aims to update the current body of evidence assessing CAR-T economic evaluations and elucidate their financial efficiency. AREAS COVERED: A systematic review of the economic evaluations of tisagenlecleucel, axicabtagene ciloleucel, idecabtagene vicleucel, lisocabtagene maraleucel, ciltacabtagene autoleucel and brexucabtagene autoleucel was performed. EXPERT OPINION: The updated results corroborated the previously reported favorable cost-effectiveness ratio of CAR-T. They also pointed out differences among CAR-T agents. However, their budget impact emerges as a significant barrier in the reimbursement process. Any proposed Managed Entry Agreement must integrate the ingrained uncertainty of long-term efficacy and precede reimbursement decisions.
Assuntos
Receptores de Antígenos Quiméricos , Humanos , Análise Custo-Benefício , Imunoterapia Adotiva , Orçamentos , Terapia Baseada em Transplante de Células e TecidosRESUMO
Relapsed/refractory central nervous system (CNS) lymphoma, whether primary or secondary, is associated with poor prognosis with currently available treatment modalities, including high-dose chemotherapy-autologous stem cell transplantation. The pivotal ZUMA-1 and JULIET trials that led to FDA approval of Axicabtagene ciloleucel and Tisagenlecleucel for relapsed refractory large cell lymphoma excluded patients with CNS involvement due to concerns of increased toxicity. However, TRANSCEND study for Lisocabtagene maraleucel in relapsed refractory large cell lymphoma allowed patients with CNS involvement and reported manageable CNS toxicities in these patients. In the real-world experience, chimeric antigen receptor T-cell (CAR T) therapy has been deemed safe and effective for these patients with poor prognosis. In this systematic review, we analyzed available literature to evaluate the role of CAR T-cell therapy in both primary and secondary CNS lymphoma using Embase, Cochrane, and PubMed databases. A total of 14 studies, including 8 retrospective analyses and 6 prospective studies/clinical trials, were included in the qualitative synthesis to study the safety and efficacy of CAR T. Based on our analysis, CAR T-cell therapy appears to be associated with reasonable efficacy and a manageable safety for primary and secondary CNS lymphoma.