echemi logo
Product
  • Product
  • Supplier
  • Inquiry
    Home > Active Ingredient News > Blood System > 【Leukemia】First-line treatment and outcome after CD19-CAR-T treatment failure for large B-cell lymphoma

    【Leukemia】First-line treatment and outcome after CD19-CAR-T treatment failure for large B-cell lymphoma

    • Last Update: 2022-11-25
    • Source: Internet
    • Author: User
    Search more information of high quality chemicals, good prices and reliable suppliers, visit www.echemi.com

    CD19-CAR-T

    CD19-CAR-T has changed the treatment landscape for relapsed/refractory large B-cell tumor lymphoma (r/r LBCL) to an unprecedented 70% high response rate, yet 60% of patients will eventually relapse or progress
    after CD19-CAR-T treatment 。 In this case, Polatuzumab, tafasitamab, celiniso, and loncastuximab have been approved by the FDA, while immune checkpoint inhibitors, lenalidomide, bispecific antibodies, investigational CAR-T products, and allogeneic hematopoietic cell transplantation, and salvage chemotherapy are additional options
    .

     

    The problem is that it is unclear how these treatments should be used after CAR-T exposure
    .
    Small studies have reported experience in treating relapse after CAR-T cell therapy, but treatment strategies vary
    .

     

    In order to report the characteristics and outcomes of LBCL patients with relapsed or progression after CD19-CAR-T therapy, to analyze remission and overall survival of first-line interventions after CAR-T therapy, to identify risk factors for adverse outcomes, and to develop a stratified model of mortality risk in patients treated after CAR-T therapy, Memorial Sloan Kettering Cancer Center Miguel-Angel Perales and Roni Shouval et al.
    conducted a retrospective observational analysis, A total of 305 patients with LBCL treated with CD19-CAR-T were enrolled in the two centers, of which 182 had disease recurrence or progression and 135 had received follow-up anti-cancer therapy
    .
    The results of the study were recently published in Leukemia
    .

     

     

    Study design

    The authors' retrospective analysis included adult (age ≥18 years) r/r LBCL patients (Figure 1A) who received CD19 CAR-T cell therapy at Memorial Sloan Kettering Cancer Center (MSKCC) and Sheba Medical Center (Israel) between April 2016 and May 2021 (Figure 1A), CD19-CAR-T products including: axicabtagene ciloleucel (axi-cel), tisagenlecleucel ( tisa-cel), lisocabtagene maraleucel (liso-cel) or POC CD28-based products (NCT02772198).

     

     

     

    Study results

    Patient characteristics and outcomes

     

    A total of 305 patients (Table 1) at both centers received CD19-CAR-T (axi-cel [n = 116, 38%]; tisa-cel [n= 83,27%]; lisocel [n= 28,9%]; POC-CAR-T [n= 78,26%]), with a median age of 63 years, the predominant histologic subtype of LBCL is non-specific diffuse large B-cell lymphoma (n = 236,77%)
    。 Most (211,69%) had previously received ≥ 3rd line therapy, 125 patients (41%) received > 3rd line therapy, most were stage III–IV (216.
    71%), and 134 (44%) and 43 (14%) had primary refractory disease and large mass tumors (the presence of any single mass > 10 cm in diameter),
    respectively.

     

     

    Grade CRS and ICANS of any grade occurred in 76% and 32% of patients, respectively, and severe (grade ≥2) CRS and ICANS
    , respectively.
    Most patients respond to CAR-T therapy (optimal ORR 67% of which 48% CR, 19% PR).

    CAR-T product-specific toxicity and mitigation are shown in the table below
    .

     


    At a median follow-up of 20 months, OS and EFS rates at 1 year after CAR-T were 62% and 29%, respectively (Figure 1B), and the cumulative recurrence or progression rate at 1 year was 63% (Figure 1C).

     

    Predictors of EFS after CAR-T

     

    The authors screened candidate predictors
    in univariate analysis.
    Subjects meeting the P<0.
    1 significance criteria were introduced into a multivariate Cox regression model (Table 2).

    The results showed that non-germinal central lymphoma (HR = 1.
    43; P = 0.
    026), primary refractory disease before apheresis (HR = 1.
    64; P = 0.
    003), elevated LDH before CAR-T (HR= 1.
    89; P< 0.
    001), and active disease during CAR-T infusion (PR HR= 1.
    90, SD/PD HR= 3.
    32; overall P = 0.
    012) were negative factors of EFS.
    In addition to these traditional risk factors, Tisa-cel also had a higher risk of treatment failure than axi-cel (HR = 2.
    03; P = 0.
    005).

     

     

    Features of the disease in the event of failure of CAR-T therapy

     

    Of the 182 patients with recurrent or persistent disease after CAR-T, 51% had CAR-T refractory disease and 49% initially responded to CAR-T but subsequently relapsed
    .
    Tissue biopsies were performed in 77 patients to confirm disease persistence or recurrence
    .
    Since antigen escape is a potential mechanism of CAR-T resistance, the authors evaluated CD19 expression
    in 52 tumor biopsy tissues.
    Interestingly, CD19 expression (Figure 2A) was only missing in 3/52 (6%), while the rest of the sample was dim (14/52, 27%) or normal (35/52, 67%) expression
    .

     

     

    The authors compared baseline characteristics of patients refractory to CAR-T with those who relapsed to identify features that may be relevant to the type of CAR-T treatment failure (Figure 2B).

    It was found that primary refractory disease before apheresis was the only pre-CAR-T factor that differed between groups: the percentage was higher in CAR-T refractory populations (58% vs.
    36%, P = 0.
    003, see red box in Figure 2B).

     

    To further clarify the difference between the patterns of response to CAR-T between the two diseases (refractory vs.
    relapse), the authors investigated additional variables
    after CAR-T infusion.
    Patients in the refractory group were more likely to develop new sites of disease involvement on disease evaluation compared with the relapsed group (14% vs.
    30%, P = 0.
    017).

    CD19 expression at disease progression in CAR-T-resistant patients is similar to that in patients with disease relapse, regardless of the expression class considered (deletion/dim/normal; P = 0.
    88) or the MFI ratio (relative to the reference value) as a continuous covariate (P = 0.
    60).

    Overall, the results of this study suggest that both drug resistance and relapse are common patterns of CAR-T treatment failure, and better biomarkers are needed to describe the biological differences
    between them.

     

    Treatment after CAR-T therapy failure

     

    Of the 182 patients with relapse or stabilization/progression after CAR-T, 135 (74%; Figure 1A) Subsequent anti-cancer therapy after CAR-T infusion, median time after CAR-T infusion was 83 days
    .
    Most patients (75/135, 56%) were treated for disease recurrence after remission of CAR-T (CR 32%, PR 24%); The remaining patients (60/135, 44%) were treated
    for unresolved CAR-T.

     

    Among patients receiving further treatment after failure of CAR-T therapy, the overall response rate was 39% (CR 20%; PR 19%)
    。 The median time from CAR-T to next-line therapy in patients with disease progression or stabilization after CAR-T is shorter than in patients who initially respond and then relapse (50 days vs.
    123 days, p< 0.
    001).

    The median follow-up time from the start of first-line therapy after CAR-T was 15 months, and the median OS and EFS after treatment were 8.
    5 and 1.
    9 months, respectively (Figure 3A), and these adverse outcomes underscore the urgent need to improve the current
    status of treatment in patients requiring anticancer therapy after CAR-T.

     

     

    First-line therapy after CAR-T uses up to 30 different treatment strategies, grouped by major drug class or drug in the table below
    .

     

     

    The main treatment classes (Figure 3B) included polatuzumab-based (n = 29), chemotherapy (n = 17; anthracyclines or platinum-based), lenalidomide-based (n = 15), radiation therapy at the affected site (ISRT) alone (n = 15), and BTK inhibitor-based (BTKi; n= 14)
    。 Treatment allocation is at the discretion of the treating physician and varies from site to site; Local disease is mainly given ISRT alone (Figure 3C).

     

    There were meaningful differences in patient characteristics between treatment strategies: more aggressive disease features in the chemotherapy group, such as large mass disease, primary refractory disease before apheresis, and lower response rates to CAR-T; Patients treated with lenalidomide were older, had more treatment lines, had better CAR-T response, and had lower pre-treatment LDH (i.
    e.
    , pre-administration of post-CAR-T therapy) compared with other treatment groups; Patients receiving polatuzumab and BTKi as the underlying strategy are typically < 60 years of age, with more than 70% of patients having elevated
    LDH before treatment.
    CR rates with systemic therapy range from 0% for chemotherapy to more than 30% since natdomide and polatuzumab-based (Figure 3D).

     

    One-year survival after treatment with the most common systemic therapy strategies ranged from 21% for BTKi and 25% for chemotherapy versus 69% for lenalidomide-based therapy (Figure 3E
    ).
    Polatuzumab-based strategies achieved an ORR of 48% (CR n= 10/29 [34%], PR n= 4/29 [14%]), but response did not translate into OS prolongation 1 year after treatment (37%)
    .
    Considering differences in OS rates after treatment, the authors performed exploratory analyses to compare major treatment strategies
    .
    In a multivariate Cox regression model that stratified by center and corrected for underlying drivers of treatment choice, including age, treatment-time LDH, and previous CAR-T response, lenalidomide-based therapy was associated with post-treatment OS improvement compared with standard chemotherapy (HR = 0.
    25, P = 0.
    027).

    Suggests that new drugs can induce remission
    after failure of CAR-T therapy.

     

    Risk stratification system after CAR-T

     

    Given the poor outcomes after first-line treatment following CAR-T treatment failure, the authors sought to identify key predictors of mortality at this time, investigating the correlation
    between disease, patient, and treatment-related features and post-treatment OS in univariate and multivariate Cox regression models 。 In univariate analysis, advanced age (> 65 years), larger lesion volume at apheresis, elevated LDH before treatment, shorter time between CAR-T and treatment (< 100 days), late stage of disease at treatment, and refractory CAR-T were candidate predictors of shorter OS after treatment (Table 3).

     

     

    A multivariate Cox model for first-line treatment of OS after CAR-T failure, including candidate predictors, confirmed independent associations
    in large mass disease (HR = 2.
    27), older age (HR = 2.
    65), elevated LDH (HR = 2.
    95), and CAR-T refractory (HR = 2.
    33).
    。 To stratify the risk of death in these patients, the authors proposed a prognostic system that included these four factors (preapheresis macromass disease, CAR-T refractory, age > 65 years, and pretreatment LDH), divided into 0-1 factors and ≥2 factors, with 1-year survival rates of 56% and 19%, respectively (Figure 3F).

     

     

    conclusion

    This study fills the knowledge gap
    related to outcomes and subsequent treatment in patients who failed CAR-T therapy with data from 182 patients with LBCL who received CD19-CAR-T therapy.
    This study also reflects the discouraging consequences of the persistence of post-CAR-T lymphoma and highlights the unmet needs
    that remain.
    Nevertheless, new drugs can show activity after CART exposure and warrant further exploration
    .
    Finally, the authors propose a prognostic tool to identify patients at highest risk of early death after CAR-T, which may be useful in stratifying patients for future studies
    after CAR-T.

     

     

    References

    Tomas AA,et al.
    Outcomes of first therapy after CD19-CAR-T treatment failure in large B-cell lymphoma.
    Leukemia .
    2022 Nov 5.
    doi: 10.
    1038/s41375-022-01739-2.

    This article is an English version of an article which is originally in the Chinese language on echemi.com and is provided for information purposes only. This website makes no representation or warranty of any kind, either expressed or implied, as to the accuracy, completeness ownership or reliability of the article or any translations thereof. If you have any concerns or complaints relating to the article, please send an email, providing a detailed description of the concern or complaint, to service@echemi.com. A staff member will contact you within 5 working days. Once verified, infringing content will be removed immediately.

    Contact Us

    The source of this page with content of products and services is from Internet, which doesn't represent ECHEMI's opinion. If you have any queries, please write to service@echemi.com. It will be replied within 5 days.

    Moreover, if you find any instances of plagiarism from the page, please send email to service@echemi.com with relevant evidence.