GLORIA

GEOMAR Library Ocean Research Information Access

Your email was sent successfully. Check your inbox.

An error occurred while sending the email. Please try again.

Proceed reservation?

Export
Filter
Material
Language
  • 1
    In: Journal of Clinical Oncology, American Society of Clinical Oncology (ASCO), Vol. 35, No. 15_suppl ( 2017-05-20), p. 7028-7028
    Abstract: 7028 Background: r/r B-ALL was reported as the most-threatening disease because of the low disease free survival even treatment with allogeneic hematopoietic stem cell transplantation. For overcoming conventional therapies limitation, autologous CD19CAR-T was performed in our clinical trials to induce remission in patients with r/r disease. 30 patients (from 7 clinical centers, in China) as volunteers with r/r B-ALL were treated by autologous CD19 CAR-T. Methods: 5 juveniles and 25 adults with r/r ALL received autologous CD19 targeted CAR-T, the doses between 1.03 × 10 6 CAR-T cells/kg and 10.09 × 10 6 CAR-T cells/kg. These 30 cases(from 7 clinical centers, in China) were treated with CAR-T cells from May. 8 2015 to January. 4 2017 (Table1). Patients were monitored for a response. Highly standardized CAR T cell preparation protocol and manageable CRS in most were kept for no significant difference in 7 clinical centers. Results: After treated with CAR-T, a total of 30 cases (5 juveniles and 25 adults coming from 7 clinical centers, in China) with r/r B-ALL were all detected the CAR-T cells proliferated in the blood and bone marrow. The results showed that complete remission (CR) is 26/30(86.67% ) between day7-14 after CD19 CART cell infusion, and 25/30(83.33%) cases arrived at MRD negative. There is about 1/3 of the total cases receiving a repeat infusions following initial ones since these patients have no safety concerns. Additionaly, the severe Cytokine release syndrome (CRS) was 8/30(26.67%) of cases and 24/30(80%) of cases was seen CRS. The anti-IL6R agent tocilizumab and Methylprednisolone were effective confrontation Severe CRS. Conclusions: This is the first multicentre report to our knowledge of successful treatment of r/r ALL with anti-CD19 CAR T cells in China. Even r/r B-ALL with high-burden leukemia patients also was effective and associated with a high remission rate after infused autologous CD19 CAR-T).(NCT 02813837).
    Type of Medium: Online Resource
    ISSN: 0732-183X , 1527-7755
    RVK:
    RVK:
    Language: English
    Publisher: American Society of Clinical Oncology (ASCO)
    Publication Date: 2017
    detail.hit.zdb_id: 2005181-5
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 2
    Online Resource
    Online Resource
    Wiley ; 2022
    In:  WIREs Mechanisms of Disease Vol. 14, No. 6 ( 2022-11)
    In: WIREs Mechanisms of Disease, Wiley, Vol. 14, No. 6 ( 2022-11)
    Abstract: Chimeric antigen receptor T‐cell (CAR‐T) treatment has revolutionized the landscape of cancer therapy with significant efficacy on hematologic malignancy, especially in relapsed and refractory B cell malignancies. However, unexpected serious toxicities such as cytokine release syndrome (CRS) and immune effector cell‐associated neurotoxicity syndrome (ICANS) still hamper its broad application. Clinical trials using CAR‐T cells targeting specific antigens on tumor cell surface have provided valuable information about the characteristics of ICANS. With unclear mechanism of ICANS after CAR‐T treatment, unremitting efforts have been devoted to further exploration. Clinical findings from patients with ICANS strongly indicated existence of overactivated peripheral immune response followed by endothelial activation‐induced blood–brain barrier (BBB) dysfunction, which triggers subsequent central nervous system (CNS) inflammation and neurotoxicity. Several animal models have been built but failed to fully replicate the whole spectrum of ICANS in human. Hopefully, novel and powerful technologies like single‐cell analysis may help decipher the precise cellular response within CNS from a different perspective when ICANS happens. Moreover, multidisciplinary cooperation among the subjects of immunology, hematology, and neurology will facilitate better understanding about the complex immune interaction between the peripheral, protective barriers, and CNS in ICANS. This review elaborates recent findings about ICANS after CAR‐T treatment from bed to bench, and discusses the potential cellular and molecular mechanisms that may promote effective management in the future. This article is categorized under: Cancer 〉 Biomedical Engineering Immune System Diseases 〉 Molecular and Cellular Physiology Neurological Diseases 〉 Molecular and Cellular Physiology
    Type of Medium: Online Resource
    ISSN: 2692-9368 , 2692-9368
    URL: Issue
    Language: English
    Publisher: Wiley
    Publication Date: 2022
    detail.hit.zdb_id: 3119452-7
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 3
    Online Resource
    Online Resource
    Springer Science and Business Media LLC ; 2021
    In:  Bone Marrow Transplantation Vol. 56, No. 12 ( 2021-12), p. 3088-3090
    In: Bone Marrow Transplantation, Springer Science and Business Media LLC, Vol. 56, No. 12 ( 2021-12), p. 3088-3090
    Type of Medium: Online Resource
    ISSN: 0268-3369 , 1476-5365
    RVK:
    RVK:
    Language: English
    Publisher: Springer Science and Business Media LLC
    Publication Date: 2021
    detail.hit.zdb_id: 2004030-1
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 4
    In: Blood, American Society of Hematology, Vol. 122, No. 21 ( 2013-11-15), p. 3482-3482
    Abstract: Gamma delta T cell, expressing gamma delta T cell receptor (gamma delta TCR), is considered as a group of unconventional T cells linking innate and adaptive immunity. Although gamma delta T cells only represent a minor population in human immune system, their functions are very broad. Recent researches have suggested that depending on the microenvironment, gamma delta T cells can assume features reminiscent of Th1, Th2, Th17 and regulatory T cells (Treg) as well as professional antigen present cells. Regulatory gamma delta T cell (gamma delta Treg) is a recently reported subset of gamma delta T cells characterized by both expressions of gamma delta TCR and forkhead/winged-helix family transcriptional repressor p3 (Foxp3), with potential immunosuppressive functions. Researches that focused on gamma delta Treg have showed the regulatory roles it has in preventing autoimmune response and relieving autoimmune diseases such as systemic lupus erythematosus (SLE), and the suppressive activity of breast tumor-derived gamma delta Tregs on innate and adaptive immunity, as well as the protective effects enhanced by gamma delta Tregs against xenogenic graft-versus-host disease in humanized mice found in our previous studies. All these observations suggest the important roles that gamma delta Treg plays in human immune system and its potential clinical applications. However, the further studies of gamma delta Treg are limited mainly due to its low quantities in vivo and the lack of methods to induce gamma delta Treg largely in vitro. It would be much significant if we found efficient induction and expansion methods for functional gamma delta Tregs. As the mammalian target of rapamycin (mTOR) is an important integrative kinase that acts as a crucial negative regulator of Treg differentiation and expansion, which can inhibit the expression of Foxp3 induced by TGF-β1, and Rapamycin (Rapa) is an immunosuppressive agent which acts through the blockade of mTOR, here we studied whether Rapa, together with TGF-β1/IL-2/IL-15, could induce and expand gamma delta Tregs derived from human peripheral blood mononuclear cells (hPBMCs) efficiently in vitro, under the stimulation of zoledronic acid (ZOL). Our results demonstrated that Rapa, synergized with TGF-β1/IL-2/IL-15, could not only facilitate the generation of gamma delta Tregs largely in vitro, with the percentage of induced gamma delta Tregs increasing from 2.4±1.2% cultured without TGF-β1/IL-15 nor Rapa, to 55.2±8.2% with Rapa and TGF-β1/IL-2/IL-15 together, compared with 26.2±6.7% with TGF-β1/IL-2/IL-15 but no Rapa (as Figure 1A indicated), but also enhanced the Foxp3 and CD25 expression levels in Rapa-induced gamma delta Tregs (as Figure 1C). Other regulatory-associated molecules expressed in Rapa-induced gamma delta Tregs included CTLA-4, ICOS and HLA-DR, and approximately no CD127, which were somehow similar with conventional CD4+CD25+ Tregs and were related with strong immunosuppressive functions. Through the co-culturing with naïve T cells derived from hPBMCs in vitro, we found that thus-induced gamma delta Tregs displayed concentration-dependent suppressive activity against the proliferation of naïve T cells, and this suppressive function appeared much greater in Rapa-induced gamma delta Tregs, compared with those induced without Rapa (as Figure 1B indicated).Figure 1Figure 1. In conclusion, our results demonstrated that Rapa, combined with TGF-β1/IL-2/IL-15, could induce and expand gamma delta Tregs largely in vitro and thus-induced gamma delta Tregs expressed high levels of Foxp3 and CD25, and displayed significant immunosuppressive activities in vitro, which provided bases to some extent for the further studies on gamma delta Treg and its potential clinical applications. Disclosures: No relevant conflicts of interest to declare.
    Type of Medium: Online Resource
    ISSN: 0006-4971 , 1528-0020
    RVK:
    RVK:
    Language: English
    Publisher: American Society of Hematology
    Publication Date: 2013
    detail.hit.zdb_id: 1468538-3
    detail.hit.zdb_id: 80069-7
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 5
    In: Cancer Biology & Medicine, China Anti-cancer Association, ( 2023-03-02), p. 129-146
    Abstract: Adoptive cellular immunotherapy with chimeric antigen receptor (CAR) T cells has emerged as a novel modality for treating relapsed and/or refractory B-cell non-Hodgkin lymphoma (B-NHL). With increasing approval of CAR T-cell products and advances in CAR T cell therapy, CAR T cells are expected to be used in a growing number of cases. However, CAR T-cell-associated toxicities can be severe or even fatal, thus compromising the survival benefit from this therapy. Standardizing and studying the clinical management of these toxicities are imperative. In contrast to other hematological malignancies, such as acute lymphoblastic leukemia and multiple myeloma, anti-CD19 CAR T-cell-associated toxicities in B-NHL have several distinctive features, most notably local cytokine-release syndrome (CRS). However, previously published guidelines have provided few specific recommendations for the grading and management of toxicities associated with CAR T-cell treatment for B-NHL. Consequently, we developed this consensus for the prevention, recognition, and management of these toxicities, on the basis of published literature regarding the management of anti-CD19 CAR T-cell-associated toxicities and the clinical experience of multiple Chinese institutions. This consensus refines a grading system and classification of CRS in B-NHL and corresponding measures for CRS management, and delineates comprehensive principles and exploratory recommendations for managing anti-CD19 CAR T-cell-associated toxicities in addition to CRS.
    Type of Medium: Online Resource
    ISSN: 2095-3941
    Language: Unknown
    Publisher: China Anti-cancer Association
    Publication Date: 2023
    detail.hit.zdb_id: 2676322-9
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 6
    Online Resource
    Online Resource
    China Science Publishing & Media Ltd. ; 2022
    In:  Journal of Zhejiang University (Medical Sciences) Vol. 51, No. 2 ( 2022-4-1), p. 192-203
    In: Journal of Zhejiang University (Medical Sciences), China Science Publishing & Media Ltd., Vol. 51, No. 2 ( 2022-4-1), p. 192-203
    Type of Medium: Online Resource
    ISSN: 1008-9292
    Language: English
    Publisher: China Science Publishing & Media Ltd.
    Publication Date: 2022
    detail.hit.zdb_id: 2992999-4
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 7
    Online Resource
    Online Resource
    Springer Science and Business Media LLC ; 2014
    In:  Annals of Hematology Vol. 93, No. 9 ( 2014-9), p. 1499-1508
    In: Annals of Hematology, Springer Science and Business Media LLC, Vol. 93, No. 9 ( 2014-9), p. 1499-1508
    Type of Medium: Online Resource
    ISSN: 0939-5555 , 1432-0584
    Language: English
    Publisher: Springer Science and Business Media LLC
    Publication Date: 2014
    detail.hit.zdb_id: 1458429-3
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 8
    In: Molecular Therapy, Elsevier BV, Vol. 29, No. 2 ( 2021-02), p. 645-657
    Type of Medium: Online Resource
    ISSN: 1525-0016
    Language: English
    Publisher: Elsevier BV
    Publication Date: 2021
    detail.hit.zdb_id: 2001818-6
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 9
    In: Journal of Immunological Methods, Elsevier BV, Vol. 402, No. 1-2 ( 2014-01), p. 82-87
    Type of Medium: Online Resource
    ISSN: 0022-1759
    RVK:
    Language: English
    Publisher: Elsevier BV
    Publication Date: 2014
    detail.hit.zdb_id: 1500495-8
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 10
    In: Blood, American Society of Hematology, Vol. 138, No. Supplement 1 ( 2021-11-05), p. 3395-3395
    Abstract: The outcome of Philadelphia chromosome-positive acute lymphoblastic leukemia (Ph+ ALL) have greatly improved in tyrosine kinase inhibitors (TKIs) era and is just moving to a chemo-free era using dasatinib and blinatumomab (Foà R, N Engl J Med. 2020; Ravandi F, Blood. 2019). However, the outcome of T315I/compound-mutated Ph+ ALL patients is dismal (Cortes JE, N Engl J Med. 2013), representing an unmet need. Recently, Scherr et al reported the curative potential of venetoclax-TKIs-dexamethasone in a BCR-ABL+ mouse model (Scherr M, Leukemia. 2019). Here, we firstly reported outcome of 19 T315I/compound-mutated relapsed/refractory (R/R)Ph+ ALL patients treated with venetoclax(100mg d1, 200mg d2, 400mg d3-28), ponatinib (45mg d1-28) and dexamethasone (0.15mg/kg d1-21,0.075mg/kg d22-28)(VPD regimen) (Figure 1A) between January 2020 and May 2021. They had already received a median of 3 lines of salvage therapy. After one cycle, 17 patients (89.5%) achieved CR/CRi [13 minimal residual disease (MRD)--negative by flow cytometry;11 major molecular remission (MMR);8 complete molecular remission (CMR)] (Figure 1B). Subsequently relapse occurred in 1/6 [allogeneic hematopoietic cell transplantation (allo-HSCT) group)] and 7/11 (VDP consolidation group). At a median follow-up of 259 days, the median EFS and OS of patients from the starting VPD treatment was 242 and 400 days. Adverse events of VPD regimen were listed in Figure 1C. Grade 3-4 neutropenia, anemia and thrombocytopenia occurred in 73.7%,36.8% and 52.6% patients. 5.3% and 16% patients have grade 3-4 rash and infection separately. No tumor lysis syndrome or death occurred. 7/19 patients were treated safety outpatient. Moreover, venetoclax had a strong synergistic effect with ponatinib and dexamethasone on inducing apoptosis of primary blast cells and BaF3 cells expressing p190 BCR/ABL with T315I-mutation in vitro, with a combination index of 0.019 when the suppressing rate is 0.05, while the effect was significant decreased when ponatinib was replaced by dasatinib (Figure 1D-F), a prominent change of mitochondrial membrane potential as well as the cleavage of PARP were also observed in triple-combination treatment group (Figure 1G-H). For T315I/compound-mutated Ph+ ALL, VPD regimen exhibited 89.5% CR/CRi rate, with deep molecular remission (57.9% MMR), while ponatinib alone showed 41% hematologic response (Cortes JE, N Engl J Med. 2013), which supported by the preclinical data suggesting TKIs and venetoclax are highly synergistic in BCR-ABL + cells in vitro (Scherr M, Leukemia. 2019). 7/11 and 1/6 patients subsequently relapsed in continuous VPD and allo-HSCT postremission treatment group separately, suggested bridging to allo-HSCT after remission is warranted. Moreover, novel compounds such as blinatumomab showed a preliminary efficacy (Couturier MA, Leuk Lymphoma. 2021). In summary, VPD regimen provides a novel treatment for T315I/compound-mutated R/R Ph+ALL under a complete oral and chemo-free model. A clinical trial also using similar VPD regimen for treatment of R/R Ph+ ALL is ongoing now (Short NJ, Am J Hematol. 2021). Figure 1 Figure 1. Disclosures No relevant conflicts of interest to declare.
    Type of Medium: Online Resource
    ISSN: 0006-4971 , 1528-0020
    RVK:
    RVK:
    Language: English
    Publisher: American Society of Hematology
    Publication Date: 2021
    detail.hit.zdb_id: 1468538-3
    detail.hit.zdb_id: 80069-7
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
Close ⊗
This website uses cookies and the analysis tool Matomo. More information can be found here...