2.Toxicity Management and Efficacy Evaluation of BCMA-CART in the Treatment of Relapsed and Refractory Multiple Myeloma.
Xiao-Yuan ZHANG ; Han-Yi DING ; Dong-Xu GANG ; Xiao-Yu HE ; Yong-Yong MA ; Hong-Lan QIAN ; Xuan-Ru LIN ; Chong-Yun XING ; Yu ZHANG ; Song-Fu JIANG
Journal of Experimental Hematology 2022;30(2):466-475
OBJECTIVE:
To investigate the toxicity management and efficacy evaluation of BCMA-chimeric antigen receptor T cells(CART) in the treatment of relapsed and refractory multiple myeloma (MM).
METHODS:
The efficacy and adverse reactions of 21 patients with MM who received BCMA-CART treatment at the First Affiliated Hospital of Wenzhou Medical University from December 2017 to September 2020 were evaluated, and the efficacy assessment and survival analysis for high-risk patients and non-high-risk patients were evaluated.
RESULTS:
After infusion of BCMA-CART cells in 21 MM patients, the number of effective cases was 17, of which the complete remission (sCR/CR) was 10, and the partial remission (VGPR/PR) was 7. The median OS time for all patients was 19.4 months, and the median PFS time was 7.9 months. The number of patients with extramedullary disease(EMD), high-risk genetics, and ISS stage Ⅲ were 5, 15 and 8, and the effective number was 3, 11 and 6, respectively. The treatment of 3 patients without high-risk factors was effective. The median OS and median PFS of patients with EMD were 14.2 and 2.5 months, respectively, which were shorter than those of patients without EMD (19.4 months and 8.9 months, respectively). The median OS and median PFS of patients with high-risk cytogenetic factors and ISS Ⅲ were not significantly different from those of non-high-risk patients. Cytokine release syndrane (CRS) occurred in 20 patients, of which 14 cases were Grade 1 CRS, while 6 were Grade 2, no CRS of Grade 3 or above occurred. IL-6 receptor inhibitors were used in 9 patients. All CRS were controlled effectively, and no patients had neurological toxicity.
CONCLUSION
BCMA-CART is a certain curative effect in the treatment of relapsed and refractory multiple myeloma, and the adverse reactions can be well controlled through close monitoring and timely treatment.
B-Cell Maturation Antigen
;
Humans
;
Immunotherapy, Adoptive/adverse effects*
;
Multiple Myeloma/therapy*
;
Receptors, Chimeric Antigen
;
Remission Induction
4.Progress in clinical studies of chimeric antigen receptor engineered T cells for treatment of childhood cancer.
Ya-Ru NI ; Xiao-Jun XU ; Yong-Min TANG
Chinese Journal of Contemporary Pediatrics 2017;19(11):1219-1224
Nowadays, the 5-year survival rate of childhood cancer patients can be more than 80%, but some patients with relapse and refractory cancers have shown no good response to traditional strategies. Chimeric antigen receptor engineered T (CAR-T) cell therapy is promising for these patients. CAR-T cells recognize the tumor-associated antigens in a non-major histocompatibility complex-restricted manner, so their anti-tumor ability is enhanced. There are four generations of CAR-T cells now. The complete remission rate of pediatric patients with relapse and refractory acute lymphoblastic leukemia can be as high as 90% when treated with CD19-targeting CAR-T cells. Furthermore, CAR-T cell therapy can also be used to bridge to transplantation and donor CAR-T cell infusion can be a strategy to prevent relapse after hematopoietic stem cell transplantation. As to solid tumors, only patients with neuroblastoma present good response to the GD2-targeting CAR-T cell therapy. The toxic or side effects of CAR-T cell therapy include cytokine release syndrome, off-tumor effect, tumor lysis syndrome, and insertion mutation. Although the CD19-targeting CAR-T cell therapy for childhood cancer can result in a high remission rate, the relapse rate is high, including CD19and CD19relapse. The mechanisms for relapse merit further investigatio.
Antigens, CD19
;
immunology
;
Child
;
Humans
;
Immunotherapy, Adoptive
;
adverse effects
;
methods
;
Neoplasms
;
therapy
;
Receptors, Antigen, T-Cell
;
genetics
;
T-Lymphocytes
;
transplantation
5.Clinical development of chimeric antigen receptor-T cell therapy for hematological malignancies.
Chinese Medical Journal 2023;136(19):2285-2296
Cellular therapies have revolutionized the treatment of hematological malignancies since their conception and rapid development. Chimeric antigen receptor (CAR)-T cell therapy is the most widely applied cellular therapy. Since the Food and Drug Administration approved two CD19-CAR-T products for clinical treatment of relapsed/refractory acute lymphoblastic leukemia and diffuse large B cell lymphoma in 2017, five more CAR-T cell products were subsequently approved for treating multiple myeloma or B cell malignancies. Moreover, clinical trials of CAR-T cell therapy for treating other hematological malignancies are ongoing. Both China and the United States have contributed significantly to the development of clinical trials. However, CAR-T cell therapy has many limitations such as a high relapse rate, adverse side effects, and restricted availability. Various methods are being implemented in clinical trials to address these issues, some of which have demonstrated promising breakthroughs. This review summarizes developments in CAR-T cell trials and advances in CAR-T cell therapy.
Humans
;
Receptors, Chimeric Antigen
;
Receptors, Antigen, T-Cell/genetics*
;
Immunotherapy, Adoptive/adverse effects*
;
Hematologic Neoplasms/therapy*
;
Multiple Myeloma/etiology*
;
Cell- and Tissue-Based Therapy
6.Secondary donor-derived CD19 CAR-T therapy is safe and efficacious in acute lymphoblastic leukemia with extramedullary relapse after first autologous CAR-T therapy.
Delin KONG ; Tingting YANG ; Jia GENG ; Ruirui JING ; Qiqi ZHANG ; Guoqing WEI ; He HUANG ; Yongxian HU
Journal of Zhejiang University. Science. B 2022;23(10):876-880
Despite the advancement of treatments, adults with relapsed/refractory (R/R) B-lineage acute lymphoblastic leukemia (B-ALL) have poor prognosis, with an expected five-year overall survival (OS) rate of 10%‒20% (Nguyen et al., 2008; Oriol et al., 2010). Extramedullary relapse of B-ALL is regarded as a high-risk factor generally associated with poor survival, occurring in about 15% to 20% of all relapsed patients (Ding et al., 2017; Sun et al., 2018). The central nervous system (CNS) and the testes are the most common sites of extramedullary relapse of B-ALL. In addition, extramedullary leukemia can appear in the skin, eyes, breasts, bones, muscles, and abdominal organs. The prognosis of relapsed extramedullary B-ALL after allogeneic hematopoietic stem cell transplantation (allo-HSCT) is extremely poor (Spyridonidis et al., 2012; Dahlberg et al., 2019). Conventional chemotherapy or radiation is often ineffective in such patients. At present, there are no optimal treatment strategies for treating extramedullary leukemia after allo-HSCT.
Adult
;
Antigens, CD19
;
Hematopoietic Stem Cell Transplantation
;
Humans
;
Immunotherapy, Adoptive/adverse effects*
;
Precursor Cell Lymphoblastic Leukemia-Lymphoma/therapy*
;
Receptors, Chimeric Antigen
;
Recurrence
7.Autologous adoptive immunotherapy without destruction of infected cells during the treatment of chronic hepatitis B.
Yan GAO ; Lai WEI ; Hong-song CHEN ; Jin SUN ; Song-xia WANG ; Yu WANG
Chinese Journal of Hepatology 2003;11(7):391-393
OBJECTIVESTo investigate the profile of liver histological damage after autologous adoptive immunotherapy with cytokine-induced killer cells (CIK cells) in patients with chronic hepatitis B (CHB).
METHODS16 CHB patients were randomly enrolled and received autologous adoptive immunotherapy, then followed up 52 weeks. Liver samples were taken from the patients to evaluate the degree of inflammation and fibrosis. The markers of hepatitis B virus and liver function were also detected.
RESULTS4 patients had two liver-biopsied samples before therapy and after 52 weeks follow-up. One patient's histological assessment revealed a significant improvement in intralobular necroinflammation (G2 --> G1) and fibrosis (S2 --> S1). The others failed to show obvious changes in liver histology. After 10 days culture in vitro, phenotypic characterization of CIK cells changed significantly by flow cytometry. The percentage of CD4+ cells decreased gradually, while the percentage of CD8+ cells increased from 20% to 60% - 80%. After 52 weeks follow-up, HBV DNA was negative (HBV DNA<4pg/ml in serum) in 6 out of 14 patients. The rates of both HBeAg/anti-HBe seroconversion and alanine aminotransferase normalization were 42.86%(6/14). There was no HBsAg/anti-HBs seroconversion. There was few severe treatment-related adverse events.
CONCLUSIONAutologous adoptive immunotherapy doesn't induce the damage of liver histology in chronic hepatitis B patients, which inhibits hepatitis B virus replication by a certain noncytotoxic mechanism.
Adolescent ; Adult ; Aged ; CD4-CD8 Ratio ; DNA, Viral ; analysis ; Hepatitis B, Chronic ; immunology ; pathology ; therapy ; Humans ; Immunotherapy, Adoptive ; adverse effects ; Liver ; pathology ; Middle Aged
8.Efficacy and safety analysis of the zanubrutinib-based bridging regimen in chimeric antigen receptor T-cell therapy for relapsed/refractory diffuse large B-cell lymphoma.
Yan LU ; Hui LIU ; Shi Guang YE ; Li Li ZHOU ; Xiu LUO ; Xiu Yong DANG ; Xiang Gui YUAN ; Wen Bin QIAN ; Ai Bin LIANG ; Ping LI
Chinese Journal of Hematology 2023;44(10):813-819
Objective: To further elucidate the clinical efficacy and safety of a combination regimen based on the BTK inhibitor zebutanil bridging CD19 Chimeric antigen receptor T cells (CAR-T cells) in the treatment of relapsed/refractory diffuse large B-cell lymphoma (r/r DLBCL) . Methods: Twenty-one patients with high-risk r/r DLBCL were treated with a zanubrutinib-based regimen bridging CAR-T between June 2020 and June 2023 at the Department of Hematology, Tongji Hospital, Tongji University and the Second Affiliated Hospital of Zhejiang University, and the efficacy and safety were retrospectively analyzed. Results: All 21 patients were enrolled, and the median age was 57 years (range: 38-76). Fourteen patients (66.7%) had an eastern cooperative oncology group performance status score (ECOG score) of ≥2. Eighteen patients (85.7%) had an international prognostic index (IPI) score of ≥3. Three patients (14.3%) had an IPI score of 2 but had extranodal infiltration. Fourteen patients (66.7%) had double-expression of DLBCL and seven (33.3%) had TP53 mutations. With a median follow-up of 24.8 (95% CI 17.0-31.6) months, the objective response rate was 81.0%, and 11 patients (52.4%) achieved complete remission. The median progression-free survival (PFS) was 12.8 months, and the median overall survival (OS) was not reached. The 1-year PFS rate was 52.4% (95% CI 29.8% -74.3%), and the 1-year OS rate was 80.1% (95% CI 58.1% -94.6%). Moreover, 18 patients (85.7%) had grade 1-2 cytokine-release syndrome, and two patients (9.5%) had grade 1 immune effector cell-associated neurotoxicity syndrome. Conclusion: Zanubrutinib-based combination bridging regimen of CAR-T therapy for r/r DLBCL has high efficacy and demonstrated a good safety profile.
Humans
;
Middle Aged
;
Receptors, Chimeric Antigen/therapeutic use*
;
Retrospective Studies
;
Immunotherapy, Adoptive/adverse effects*
;
Lymphoma, Large B-Cell, Diffuse/drug therapy*
;
Cell- and Tissue-Based Therapy
;
Antigens, CD19/adverse effects*
9.New strategy of cancer immunotherapy: irradiation or chemotherapeutics-induced lymphopenia combined with immune reconstitution and tumor vaccine.
Jun MA ; Yi-li WANG ; Hong-ming HU ; Bernard A FOX ; Lü-sheng SI
Chinese Journal of Oncology 2005;27(8):452-456
OBJECTIVETo test whether vaccination performed during irradiation or chemotherapeutics-induced lymphopenia-driven T cell proliferation could augment the antitumor immunity.
METHODSThe study composed of two parts, investigating the anti-tumor efficacy of performing tumor vaccination during early immune reconstitution period following sublethal total body irradiation and cyclophosphamide (Cy)-induced lymphopenia, respectively. Mice were subsequently reconstituted with naïve splenocytes from syngeneic mice and were named RLM (Reconstituted lymphopenic mice). Immunization/vaccination (F10) and adoptive immunotherapy (D5-G6) were used to explore anti-tumor immune responses in vaccinated irradiation/RLM and vaccinated Cy/RLM, respectively. Both normal C57BL/6 mice and RLM were vaccinated with irradiated, weakly immunogenic F10 melanoma cells and subsequently challenged with F10 cells. In addition, to determine the role of CD4(+) and CD8(+) T cells in the protective anti-tumor immune response, irradiation/RLM were depleted of these subpopulations by administration of the appropriate mAb around challenge. In the second part, adoptive immunotherapy was used to evaluate the anti-tumor immune responses under chemotherapeutics-induced lymphopenic condition. Both normal mice and RLM (Cy-treated) were vaccinated with GM-CSF-modified D5 melanoma cells (D5-G6) and tumor vaccine draining lymph nodes (TVDLN) were harvested 9-10 days later. Effector T cells were generated in vitro from TVDLN cells and adoptively transferred to mice bearing 3-day pre-established pulmonary metastases (D5). Recipient mice were sacrificed 2 weeks later after tumor inoculation and pulmonary metastases were enumerated.
RESULTSSignificantly greater protection was induced in vaccinated irradiation/RLM, compared to vaccinated normal mice (63.2% vs 16.7%). Protective immunity in RLM depended on CD8(+) T cells. Increase in the interval between reconstitution and vaccination significantly decrease the protection. Effector T cells generated from vaccinated Cy-treated RLM demonstrated significantly higher in vivo anti-tumor efficacy over those of vaccinated normal mice.
CONCLUSIONThis study suggests that vaccination of RLM could elicit augmented antitumor immunity compared to normal hosts, highlighting the potential clinical benefit of performing tumor vaccination during irradiation or chemotherapeutics-induced lymphopenia in cancer patients.
Animals ; CD8-Positive T-Lymphocytes ; immunology ; Cancer Vaccines ; therapeutic use ; Cyclophosphamide ; adverse effects ; Female ; Granulocyte-Macrophage Colony-Stimulating Factor ; immunology ; Immunotherapy, Adoptive ; methods ; Lymphopenia ; etiology ; therapy ; Melanoma, Experimental ; drug therapy ; immunology ; radiotherapy ; Mice ; Mice, Inbred C57BL ; Whole-Body Irradiation
10.Epstein Barr virus-associated lymphoproliferative diseases: the virus as a therapeutic target.
Experimental & Molecular Medicine 2015;47(1):e136-
Epstein Barr virus (EBV)-associated lymphoproliferative diseases (LPDs) express all EBV latent antigens (type III latency) in immunodeficient patients and limited antigens (type I and II latencies) in immunocompetent patients. Post-transplantation lymphoproliferative disease (PTLD) is the prototype exhibiting type III EBV latency. Although EBV antigens are highly immunogenic, PTLD cell proliferation remains unchecked because of the underlying immunosuppression. The restoration of anti-EBV immunity by EBV-specific T cells of either autologous or allogeneic origin has been shown to be safe and effective in PTLDs. Cellular therapy can be improved by establishing a bank of human leukocyte antigen-characterized allogeneic EBV-specific T cells. In EBV+ LPDs exhibiting type I and II latencies, the use of EBV-specific T cells is more limited, although the safety and efficacy of this therapy have also been demonstrated. The therapeutic role of EBV-specific T cells in EBV+ LPDs needs to be critically reappraised with the advent of monoclonal antibodies and other targeted therapy. Another strategy involves the use of epigenetic approaches to induce EBV to undergo lytic proliferation when expression of the viral thymidine kinase renders host tumor cells susceptible to the cytotoxic effects of ganciclovir. Finally, the prophylactic use of antiviral drugs to prevent EBV reactivation may decrease the occurrence of EBV+ LPDs.
Antiviral Agents/therapeutic use
;
Cell- and Tissue-Based Therapy
;
DNA Methylation
;
Epstein-Barr Virus Infections/*complications
;
Genome, Viral
;
Hematopoietic Stem Cell Transplantation
;
Herpesvirus 4, Human/*physiology
;
Humans
;
Immunotherapy, Adoptive
;
Lymphoproliferative Disorders/diagnosis/*etiology/*therapy
;
Organ Transplantation/adverse effects
;
T-Lymphocytes/immunology
;
Transplantation, Homologous
;
Virus Latency