Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 252
Filter
1.
Biomed Pharmacother ; 178: 117133, 2024 Sep.
Article in English | MEDLINE | ID: mdl-39024837

ABSTRACT

Primary central nervous system lymphoma (PCNSL) is a group of malignant brain tumors with a poor prognosis, and new therapeutic approaches for this tumor urgently need to be investigated. Formulated from a long-standing anti-inflammatory drugs, ACT001 has demonstrated in clinical research to be able to pass through the blood-brain barrier (BBB) and affect the central nervous system. The effects of ACT001 on PCNSL cell apoptosis, proliferation and immune-related indexes were detected by flow cytometry, and the efficacy of ACT001 was verified in vivo by constructing a mouse PCNSL tumor model. ACT001 significantly inhibited PCNSL cell proliferation and induced apoptosis in vitro. In addition, ACT001 can significantly inhibit the PD-1/PD-L1 expression and restore the function of T cells, so that the immune system cannot allow tumor cells to escape. In vivo experiments show that co-infusion of ACT001 and T cells effectively inhibits PCNSL tumor growth in NSG mice. Our work describes the inhibitory effect of ACT001 on the PCNSL cell line and demonstrated the inhibitory effect of ACT001 on immune checkpoints.


Subject(s)
Apoptosis , Cell Proliferation , Central Nervous System Neoplasms , Lymphoma , T-Lymphocytes , Animals , Cell Line, Tumor , Cell Proliferation/drug effects , T-Lymphocytes/drug effects , T-Lymphocytes/immunology , Mice , Apoptosis/drug effects , Central Nervous System Neoplasms/drug therapy , Central Nervous System Neoplasms/pathology , Central Nervous System Neoplasms/immunology , Lymphoma/drug therapy , Lymphoma/pathology , Lymphoma/immunology , Humans , B7-H1 Antigen/metabolism , Mice, Inbred NOD , Programmed Cell Death 1 Receptor/antagonists & inhibitors , Antineoplastic Agents/pharmacology , Xenograft Model Antitumor Assays
2.
J Neurooncol ; 169(1): 51-60, 2024 Aug.
Article in English | MEDLINE | ID: mdl-38865013

ABSTRACT

PURPOSE: Immunosuppression is a well-established risk factor for primary central nervous system lymphomas (PCNSLs), which present in this context distinct radiological characteristics. Our aim was to describe the radiological evolution of treated PCNSL in immunocompromised patients and suggest adapted MRI response criteria. METHODS: We conducted a multicenter retrospective study of patients from the French LOC, K-Virogref and CANCERVIH network databases and enrolled adult immunocompromised patients with newly diagnosed PCNSL. RESULTS: We evaluated the baseline, intermediate, end-of-treatment and follow-up MRI data of 31 patients (9 living with HIV, 16 with solid organ transplantation and 6 with an autoimmune disease under chronic immunosuppressive therapy). At baseline, 23/30 (77%) patients had necrotic lesions with ring enhancement and 28% of the lesions were hemorrhagic. At the end of the first-line treatment, 12/28 (43%) patients could not be classified according to the IPCG criteria. Thirteen of 28 (46%) patients still harbored contrast enhancement, and 11/28 (39%) patients had persistent large necrotic lesions with a median diameter of 15 mm. These aspects were not associated with a pejorative outcome and progressively diminished during follow-up. Six patients relapsed; however, we failed to identify any neuroimaging risk factors on the end-of-treatment MRI. CONCLUSION: In immunocompromised patients, PCNSLs often harbor alarming features on end-of-treatment MRI, with persistent contrast-enhanced lesions frequently observed. However, these aspects seemed to be related to the necrotic and hemorrhagic nature of the lesions and were not predictive of a pejorative outcome. Specific response criteria for this population are thereby proposed.


Subject(s)
Central Nervous System Neoplasms , Immunocompromised Host , Lymphoma , Magnetic Resonance Imaging , Humans , Male , Female , Middle Aged , Central Nervous System Neoplasms/immunology , Central Nervous System Neoplasms/pathology , Central Nervous System Neoplasms/diagnostic imaging , Retrospective Studies , Immunocompromised Host/immunology , Aged , Adult , Lymphoma/diagnostic imaging , Lymphoma/pathology , Lymphoma/immunology , Follow-Up Studies
3.
Clin Cancer Res ; 30(16): 3578-3591, 2024 Aug 15.
Article in English | MEDLINE | ID: mdl-38864848

ABSTRACT

PURPOSE: Retinoblastoma is the most common intraocular malignancy in children. Although new chemotherapeutic approaches have improved ocular salvage rates, novel therapies are required for patients with refractory intraocular and metastatic disease. Chimeric antigen receptor (CAR) T cells targeting glypican-2 (GPC2) are a potential new therapeutic strategy. EXPERIMENTAL DESIGN: GPC2 expression and its regulation by the E2F1 transcription factor were studied in retinoblastoma patient samples and cellular models. In vitro, we performed functional studies comparing GPC2 CAR T cells with different costimulatory domains (4-1BB and CD28). In vivo, the efficacy of local and systemic administration of GPC2 CAR T cells was evaluated in intraocular and leptomeningeal human retinoblastoma xenograft models. RESULTS: Retinoblastoma tumors, but not healthy retinal tissues, expressed cell surface GPC2, and this tumor-specific expression was driven by E2F1. GPC2-directed CARs with 4-1BB costimulation (GPC2.BBz) were superior to CARs with CD28 stimulatory domains (GPC2.28z), efficiently inducing retinoblastoma cell cytotoxicity and enhancing T-cell proliferation and polyfunctionality. In vivo, GPC2.BBz CARs had enhanced persistence, which led to significant tumor regression compared with either control CD19 or GPC2.28z CARs. In intraocular models, GPC2.BBz CAR T cells efficiently trafficked to tumor-bearing eyes after intravitreal or systemic infusions, significantly prolonging ocular survival. In central nervous system (CNS) retinoblastoma models, intraventricular or systemically administered GPC2.BBz CAR T cells were activated in retinoblastoma-involved CNS tissues, resulting in robust tumor regression with substantially extended overall mouse survival. CONCLUSIONS: GPC2-directed CAR T cells are effective against intraocular and CNS metastatic retinoblastomas.


Subject(s)
Glypicans , Immunotherapy, Adoptive , Receptors, Chimeric Antigen , Retinoblastoma , T-Lymphocytes , Xenograft Model Antitumor Assays , Humans , Animals , Retinoblastoma/immunology , Retinoblastoma/pathology , Retinoblastoma/therapy , Mice , Receptors, Chimeric Antigen/immunology , Glypicans/immunology , Glypicans/antagonists & inhibitors , Immunotherapy, Adoptive/methods , T-Lymphocytes/immunology , T-Lymphocytes/metabolism , Cell Line, Tumor , Central Nervous System Neoplasms/therapy , Central Nervous System Neoplasms/immunology , Central Nervous System Neoplasms/secondary , Central Nervous System Neoplasms/pathology , Disease Models, Animal , Female
4.
J Clin Oncol ; 42(23): 2769-2779, 2024 Aug 10.
Article in English | MEDLINE | ID: mdl-38771986

ABSTRACT

PURPOSE: T cells modified with chimeric antigen receptors (CARTs) have demonstrated efficacy for hematologic malignancies; however, benefit for patients with CNS tumors has been limited. To enhance T cell activity against GD2+ CNS malignancies, we modified GD2-directed CART cells (GD2.CARTs) with a constitutively active interleukin (IL)-7 receptor (C7R-GD2.CARTs). METHODS: Patients age 1-21 years with H3K27-altered diffuse midline glioma (DMG) or other recurrent GD2-expressing CNS tumors were eligible for this phase I trial (ClinicalTrials.gov identifier: NCT04099797). All subjects received standard-of-care adjuvant radiation therapy or chemotherapy before study enrollment. The first treatment cohort received GD2.CARTs alone (1 × 107 cells/m2), and subsequent cohorts received C7R-GD2.CARTs at two dose levels (1 × 107 cells/m2; 3 × 107 cells/m2). Standard lymphodepletion with cyclophosphamide and fludarabine was included at all dose levels. RESULTS: Eleven patients (age 4-18 years) received therapy without dose-limiting toxicity. The GD2.CART cohort did not experience toxicity, but had disease progression after brief improvement of residual neurologic deficits (≤3 weeks). The C7R-GD2.CART cohort developed grade 1 tumor inflammation-associated neurotoxicity in seven of eight (88%) cases, controllable with anakinra. Cytokine release syndrome was observed in six of eight (75%, grade 1 in all but one patient) and associated with increased circulating IL-6 and IP-10 (P < .05). Patients receiving C7R-GD2.CARTs experienced temporary improvement from baseline neurologic deficits (range, 2 to >12 months), and seven of eight (88%) remained eligible for additional treatment cycles (range 2-4 cycles). Partial responses by iRANO criteria were observed in two of seven (29%) patients with DMG treated by C7R-GD2.CARTs. CONCLUSION: Intravenous GD2.CARTs with and without C7R were well tolerated. Patients treated with C7R-GD2.CARTs exhibited transient improvement of neurologic deficits and increased circulating cytokines/chemokines. Treatment with C7R-GD2.CARTs represents a novel approach warranting further investigation for children with these incurable CNS cancers.


Subject(s)
Central Nervous System Neoplasms , Gangliosides , Receptors, Chimeric Antigen , Humans , Child , Adolescent , Child, Preschool , Male , Female , Receptors, Chimeric Antigen/immunology , Receptors, Chimeric Antigen/therapeutic use , Infant , Young Adult , Central Nervous System Neoplasms/drug therapy , Central Nervous System Neoplasms/immunology , Central Nervous System Neoplasms/therapy , Gangliosides/immunology , Immunotherapy, Adoptive/adverse effects , Immunotherapy, Adoptive/methods , Neoplasm Grading , Glioma/drug therapy , Glioma/pathology , Glioma/therapy , Glioma/immunology , Interleukin-7 Receptor alpha Subunit
5.
J Neurooncol ; 168(3): 487-494, 2024 Jul.
Article in English | MEDLINE | ID: mdl-38658464

ABSTRACT

PURPOSE: The systemic inflammation response index (SIRI) and systemic immune-inflammation index (SII) are based on neutrophil, monocyte, platelet, and lymphocyte counts. The SIRI and SII are used to predict the survival of patients with malignant tumors. It is well known that the inflammatory immune response is closely related to cancer occurrence and progression. In the present study, we evaluated the potential prognostic significance of SIRI and SII in patients with primary central nervous system lymphoma (PCNSL). METHODS: Fifty-eight consecutive patients were enrolled in this study between November 2006 and May 2022. Among the 58 patients, 47 patients with sufficient blood test data and follow-up were analyzed. The patients with steroid intake at the time point of the blood test and higher C-reactive protein were excluded. RESULTS: The median follow-up and survival times were 31 and 36 months, respectively. The optimal cutoff SIRI value was based on the receiver operating characteristic curve (ROC) for overall survival (OS) and stratified patients into low (< 1.43 × 109/L, n = 22) and high (≥ 1.43 × 109/L, n = 25) SIRI groups. The optimal cutoff SII value based on the ROC for OS stratified patients into low (< 694.9, n = 28) and high (≥ 694.9, n = 19) SII groups. A low SIRI value was associated with longer OS (p = 0.006). Furthermore, a low SII value was associated with longer OS (p = 0.044). The prognostic factors associated with prolonged survival in univariate analysis using the Cox proportional hazard model were age < 65 years, low SIRI, and low SII. The multivariate analysis demonstrated that age < 65 years and low SIRI independently predicted longer OS. CONCLUSION: Simple, less expensive, and routinely ordered preoperative blood count assessments such as SIRI and SII predict the OS of patients with PCNSL. This study demonstrated that PCNSL is associated with pre-treatment systemic immune-inflammation states.


Subject(s)
Central Nervous System Neoplasms , Inflammation , Lymphoma , Humans , Male , Female , Middle Aged , Prognosis , Aged , Central Nervous System Neoplasms/immunology , Central Nervous System Neoplasms/mortality , Central Nervous System Neoplasms/blood , Adult , Inflammation/immunology , Inflammation/blood , Lymphoma/immunology , Lymphoma/mortality , Lymphoma/blood , Follow-Up Studies , Aged, 80 and over , Retrospective Studies , Survival Rate , Young Adult , ROC Curve , Neutrophils/immunology
6.
J Clin Neurosci ; 124: 36-46, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38642434

ABSTRACT

Primary central nervous system lymphoma (PCNSL) is a rare extranodal non-Hodgkin lymphoma, and there is limited research on its tumor microenvironment (TME). Nevertheless, more and more studies have evidence that TME has essential effects on tumor cell proliferation, immune escape, and drug resistance. Thus, it is critical to elucidate the role of TME in PCNSL. The understanding of the PCNSL TME is gradually unfolding, including factors that distinguish it from systemic diffuse large B-cell lymphoma (DLBCL). The TME in PCNSL exhibits both transcriptional and spatial intratumor heterogeneity. Cellular interactions between tumor cells and stroma cells reveal immune evasion signaling. The comparative analysis between PCNSL and DLBCL suggests that PCNSL is more likely to be an immunologically deficient tumor. In PCNSL, T cell exhaustion and downregulation of macrophage immune function are accompanied by suppressive microenvironmental factors such as M2 polarized macrophages, endothelin B receptor, HLA depletion, PD-L1, and TIM-3. MMP-9, Integrin-ß1, and ICAM-1/LFA-1 play crucial roles in transendothelial migration towards the CNS, while CXCL13/CXCR5, CD44, MAG, and IL-8 are essential for brain parenchymal invasion. Further, macrophages, YKL-40, CD31, CD105, PD-1/PD-L1 axis, osteopontin, galectin-3, aggregative perivascular tumor cells, and HLA deletion may contribute to poor outcomes in patients with PCNSL. This article reviews the effect of various components of TME on the progression and prognosis of PCNSL patients to identify novel therapeutic targets.


Subject(s)
Central Nervous System Neoplasms , Tumor Microenvironment , Humans , Tumor Microenvironment/immunology , Tumor Microenvironment/physiology , Central Nervous System Neoplasms/pathology , Central Nervous System Neoplasms/genetics , Central Nervous System Neoplasms/metabolism , Central Nervous System Neoplasms/immunology , Prognosis , Lymphoma, Non-Hodgkin/pathology
7.
Blood ; 144(4): 457-461, 2024 Jul 25.
Article in English | MEDLINE | ID: mdl-38484137

ABSTRACT

ABSTRACT: Although CD20×CD3 bispecific antibodies are effective against systemic B-cell lymphomas, their efficacy in central nervous system (CNS) lymphoma is unknown. Here, we report the CD20×CD3 bispecific glofitamab penetrates the blood-brain barrier, stimulates immune-cell infiltration of CNS tumors, and induces clinical responses in patients with secondary CNS.


Subject(s)
Antibodies, Bispecific , Central Nervous System Neoplasms , Humans , Central Nervous System Neoplasms/immunology , Central Nervous System Neoplasms/pathology , Central Nervous System Neoplasms/drug therapy , Antibodies, Bispecific/therapeutic use , Blood-Brain Barrier/pathology , Antigens, CD20/immunology , CD3 Complex/immunology , Female , Male , Lymphoma, B-Cell/immunology , Lymphoma, B-Cell/pathology , Lymphoma, B-Cell/drug therapy , Lymphoma/immunology , Lymphoma/pathology , Lymphoma/drug therapy , Middle Aged
8.
J Neurochem ; 168(6): 1157-1167, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38332527

ABSTRACT

For CNS lymphomas (CNSL), there is a high need for minimally invasive and easily obtainable diagnostic markers. Intrathecal IgM synthesis can easily be determined in routine CSF diagnostics. The aim of this study was to systematically investigate the diagnostic potential of intrathecal IgM synthesis in primary and secondary CNSL (PCNSL and SCNSL). In this retrospective study, patients with a biopsy-proven diagnosis of PCNSL or SCNSL were compared with patients with other neurological diseases in whom CNSL was initially the primary radiological differential diagnosis based on MRI. Sensitivity and specificity of intrathecal IgM synthesis were calculated using receiver operating characteristic curves. Seventy patients with CNSL were included (49 PCNSL and 21 SCNSL) and compared to 70 control patients. The sensitivity and specificity for the diagnosis of CNSL were 49% and 87%, respectively, for the entire patient population and 66% and 91% after selection for cases with tumor access to the CSF system and isolated intrathecal IgM synthesis. In cases with MRI-based radiological suspicion of CNSL, intrathecal IgM synthesis has good specificity but limited sensitivity. Because of its low-threshold availability, analysis of intrathecal IgM synthesis has the potential to lead to higher diagnostic accuracy, especially in resource-limited settings, and deserves further study.


Subject(s)
Central Nervous System Neoplasms , Immunoglobulin M , Lymphoma , Humans , Immunoglobulin M/cerebrospinal fluid , Male , Female , Middle Aged , Retrospective Studies , Central Nervous System Neoplasms/diagnosis , Central Nervous System Neoplasms/cerebrospinal fluid , Central Nervous System Neoplasms/immunology , Aged , Lymphoma/cerebrospinal fluid , Lymphoma/diagnosis , Adult , Biomarkers, Tumor/cerebrospinal fluid , Magnetic Resonance Imaging , Aged, 80 and over , Sensitivity and Specificity , Young Adult
9.
Clin Lymphoma Myeloma Leuk ; 24(4): e142-e151, 2024 Apr.
Article in English | MEDLINE | ID: mdl-38267353

ABSTRACT

Chimeric Antigen Receptor T-cell (CAR T-cell) therapy is an effective treatment for relapsed/refractory (R/R) large B cell lymphoma (LBCL). However, patients with central nervous system (CNS) lymphoma were excluded in most of the CAR T-cell therapy trials. This meta-analysis assesses the efficacy with CAR T-cell therapy in LBCL patients with CNS involvement. Two reviewers independently searched PubMed and Cochrane Library to identify all published literature associated with United States Food and Drug Administration approved CAR T-cell therapies for LBCL. Patients with CNS LBCL were included. Meta-analysis of proportion was performed to evaluate the overall response (ORR), complete response (CR) for efficacy, and cytokine release syndrome and immune effector cell-associated neurotoxicity syndrome for safety assessment. Nineteen studies were qualified for inclusion with 141 CNS LBCL patients. The ORR and CR rates were 61% and 55% respectively. The median overall survival (OS) was 8.8 months, and the median progression free survival (PFS) was 4.4 months. Severe immune effector cell-associated neurotoxicity syndrome (grade≥3) were reported in 25% (32/130) patients and severe cytokine release syndrome (grade≥3) were found in 10% (13/124) of the patients. The safety and efficacy of CAR T-cell therapy in CNS LBCL patients appears comparable to patients without CNS involvement.


Subject(s)
Central Nervous System Neoplasms , Immunotherapy, Adoptive , Lymphoma, Large B-Cell, Diffuse , Humans , Immunotherapy, Adoptive/methods , Immunotherapy, Adoptive/adverse effects , Lymphoma, Large B-Cell, Diffuse/therapy , Lymphoma, Large B-Cell, Diffuse/immunology , Central Nervous System Neoplasms/therapy , Central Nervous System Neoplasms/immunology , Central Nervous System Neoplasms/mortality , Receptors, Chimeric Antigen , Treatment Outcome , Male
10.
J Neurosurg ; 140(6): 1549-1557, 2024 Jun 01.
Article in English | MEDLINE | ID: mdl-38157532

ABSTRACT

OBJECTIVE: Malignancies of the CNS are difficult to treat because the blood-brain barrier (BBB) prevents most therapeutics from reaching the intracranial lesions at sufficiently high concentrations. This also applies to chimeric antigen receptor (CAR) T cells, for which systemic delivery is inferior to direct intratumoral or intraventricular injection of the cells. The authors previously reported on a novel approach to safely and reversibly open the BBB of mice by applying intra-arterial (IA) injections of NEO100, a pharmaceutical-grade version of the natural monoterpene perillyl alcohol. The authors hypothesized that this method would enable enhanced brain entry and therapeutic activity of intravenously delivered CAR T cells, which the authors tested in a mouse model of CNS lymphoma. METHODS: Human Raji lymphoma cells were implanted into the brains of immune-deficient mice. After tumor uptake was confirmed with bioluminescent imaging, 0.3% NEO100 was injected intra-arterially, which was followed by intravenous (IV) delivery of CD19-targeted CAR T cells. After this single intervention, tumor growth was monitored with imaging, long-term survival of mice was recorded, and select mice were euthanized to analyze the distribution of CAR T cells in brain tissue. RESULTS: Intravenously injected CAR T cells could be readily detected in brain tumor areas after IA injection of NEO100 but not after IA injection of the vehicle (without NEO100). Although all untreated control animals died within 3 weeks, all mice that received IA NEO100 followed by IV CAR T cells survived and thrived for 200 days, when the experiment was terminated. Of the mice that received IV CAR T cells without prior IA NEO100, 3 died within 3 weeks and 2 survived long-term. CONCLUSIONS: BBB opening by IA NEO100 facilitates brain entry of intravenously delivered CD19 CAR T cells. The long-term survival of all mice with CNS lymphoma, along with the disappearance of the tumor as determined with imaging, suggests that this one-time therapeutic intervention was curative. BBB opening by IA NEO100 may offer a novel option to increase brain access by CAR T cells.


Subject(s)
Immunotherapy, Adoptive , Injections, Intra-Arterial , Receptors, Chimeric Antigen , Animals , Mice , Immunotherapy, Adoptive/methods , Disease Models, Animal , Blood-Brain Barrier , Humans , Brain Neoplasms/therapy , Brain Neoplasms/pathology , Brain Neoplasms/immunology , Cell Line, Tumor/transplantation , Lymphoma/therapy , Lymphoma/immunology , Central Nervous System Neoplasms/therapy , Central Nervous System Neoplasms/immunology , T-Lymphocytes/immunology , T-Lymphocytes/transplantation , Mice, SCID
11.
J Neuroimmunol ; 382: 578156, 2023 09 15.
Article in English | MEDLINE | ID: mdl-37556888

ABSTRACT

We reported a 61-year-old man presented with 10-month progressing left sciatic neuropathy and 10-day right facial neuropathy. Serum amphiphysin-IgG was positive. 18F-FDG PET/CT of the whole body showed no signs of malignancy. Treatment with plasma exchange and oral prednisone relieved the symptoms. Nine months later, right hemiparesis and seizure of right limbs developed. 18F-FDG and 18F-PBR06 (18 kDa translocator protein, TSPO) radioligand PET/MRI of the whole body revealed intense uptake in the intracranial lesions. Intracranial lymphoma was diagnosed by stereotactic needle brain biopsy. Mononeuropathies could be paraneoplastic syndromes. TSPO shows high uptake in intracranial lymphoma on 18F-PBR06 PET images.


Subject(s)
Central Nervous System Neoplasms , Facial Nerve Diseases , Lymphoma , Sciatic Neuropathy , Humans , Male , Middle Aged , Brain/immunology , Facial Nerve Diseases/etiology , Facial Nerve Diseases/immunology , Facial Nerve Diseases/therapy , Fluorodeoxyglucose F18 , Immunoglobulin G/immunology , Positron Emission Tomography Computed Tomography/methods , Positron-Emission Tomography/methods , Radiopharmaceuticals , Receptors, GABA/metabolism , Sciatic Neuropathy/etiology , Sciatic Neuropathy/immunology , Sciatic Neuropathy/therapy , Central Nervous System Neoplasms/complications , Central Nervous System Neoplasms/diagnostic imaging , Central Nervous System Neoplasms/immunology , Autoimmune Diseases/etiology , Autoimmune Diseases/immunology , Lymphoma/complications , Lymphoma/diagnostic imaging , Lymphoma/immunology , Paraneoplastic Polyneuropathy/etiology , Paraneoplastic Polyneuropathy/immunology , Prednisone/therapeutic use , Glucocorticoids/therapeutic use , Plasma Exchange , Nerve Tissue Proteins/immunology
12.
Neuro Oncol ; 24(8): 1246-1258, 2022 08 01.
Article in English | MEDLINE | ID: mdl-35137206

ABSTRACT

BACKGROUND: CNS germ cell tumors (GCTs) predominantly develop in pediatric and young adult patients with variable responses to surgery, radiation, and chemotherapy. This study aimed to examine the complex and largely unknown pathogenesis of CNS GCTs. METHODS: We used a combined transcriptomic and methylomic approach in 84 cases and conducted an integrative analysis of the normal cells undergoing embryogenesis and testicular GCTs. RESULTS: Genome-wide transcriptome analysis in CNS GCTs indicated that germinoma had a transcriptomic profile representative of primitive cells during early embryogenesis with high meiosis/mitosis potentials, while nongerminomatous GCTs (NGGCTs) had differentiated phenotypes oriented toward tissue formation and organogenesis. Co-analysis with the transcriptome of human embryonic cells revealed that germinomas had expression profiles similar to those of primordial germ cells, while the expression profiles of NGGCTs were similar to those of embryonic stem cells. Some germinoma cases were characterized by extensive immune-cell infiltration and high expression of cancer-testis antigens. NGGCTs had significantly higher immune-cell infiltration, characterized by immune-suppression phenotype. CNS and testicular GCTs (TGCTs) had similar mutational profiles; TGCTs showed enhanced copy number alterations. Methylation analysis clustered germinoma/seminoma and nongerminoma/nonseminoma separately. Germinoma and seminoma were co-categorized based on the degree of the tumor microenvironment balance. CONCLUSIONS: These results suggested that the pathophysiology of GCTs was less dependent on their site of origin and more dependent on the state of differentiation as well as on the tumor microenvironment balance. This study revealed distinct biological properties of GCTs, which will hopefully lead to future treatment development.


Subject(s)
Central Nervous System Neoplasms , Epigenome , Neoplasms, Germ Cell and Embryonal , Transcriptome , Central Nervous System Neoplasms/genetics , Central Nervous System Neoplasms/immunology , Central Nervous System Neoplasms/pathology , Child , Embryonic Development , Germinoma/genetics , Germinoma/immunology , Humans , Male , Mutation , Neoplasms, Germ Cell and Embryonal/genetics , Neoplasms, Germ Cell and Embryonal/immunology , Neoplasms, Germ Cell and Embryonal/pathology , Seminoma/genetics , Testicular Neoplasms/genetics , Tumor Microenvironment , Young Adult
13.
Ann Clin Lab Sci ; 51(6): 790-794, 2021 Nov.
Article in English | MEDLINE | ID: mdl-34921032

ABSTRACT

OBJECTIVE: To investigate the cytokine release syndrome (CRS) condition for central nervous system B-cell acute lymphocytic leukemia (CNS B-ALL) patients after CAR-Ts targeting CD19 with short hairpin RNA (shRNA)-IL-6 gene silencing technology (ssCART-19s) infusion. METHODS: This prospective observational research included a total of 12 cases of patients with CNS B-ALL from March 2017 to February 2020. ssCART-19 infusions (5×106 cells/kg) were given to patients for 3 consecutive days. After infusion, the temperature of all patients was detected constantly and the CRS was carefully monitored within 1 month after treatment. The serum levels of IL-2, IL-4, IL-6, IL-10, IFN-γ, TNF-α, CRP and IL-17A were tested by enzyme-linked immunosorbent assay (ELISA) within 10 days after infusion. RESULTS: All 12 CNS B-ALL patients showed CRS with 100% incidence rate, with 3 cases (25.00%) of CRS stage I and 9 cases (75.00%) with CRS stage II. No CRS stage III~V was observed. The overall response rate was 91.67% (11/12), with 10 patients (83.33%) showed CR and 1 case (8.33%) of PR. In 9 patients with CRS stage II, the temperature increased persistently, ranging from 4 days to 14 days after infusion, and decreased gradually after 14 days of nursing treatment. The hyperthermia condition started from 1 day after infusion and returned to baseline at the following 2-10 days of nursing treatment. The levels of the inflammatory factors increased markedly after ssCAR-T19s infusion for 2-3 days compared to the baseline, and gradually returned to the baseline after treatment. After 10 days of infusion, all inflammatory factors returned to normal levels. CONCLUSION: ssCART-19s infusion induced short-term slight CRS with increased temperature and inflammatory factors, and no severe CRS was observed.


Subject(s)
Central Nervous System Neoplasms/therapy , Cytokine Release Syndrome , Gene Knockdown Techniques/methods , Immunotherapy, Adoptive , Interleukin-6 , Precursor Cell Lymphoblastic Leukemia-Lymphoma/therapy , Receptors, Chimeric Antigen , Adult , Antigens, CD19/immunology , Central Nervous System Neoplasms/immunology , Central Nervous System Neoplasms/pathology , Cytokine Release Syndrome/etiology , Cytokine Release Syndrome/immunology , Cytokine Release Syndrome/physiopathology , Cytokine Release Syndrome/therapy , Female , Humans , Immunotherapy, Adoptive/adverse effects , Immunotherapy, Adoptive/methods , Interleukin-6/genetics , Interleukin-6/immunology , Male , Monitoring, Physiologic/methods , Neurotoxicity Syndromes/etiology , Neurotoxicity Syndromes/immunology , Precursor Cell Lymphoblastic Leukemia-Lymphoma/immunology , Precursor Cell Lymphoblastic Leukemia-Lymphoma/pathology , RNA, Small Interfering/therapeutic use , Receptors, Chimeric Antigen/administration & dosage , Receptors, Chimeric Antigen/immunology , Treatment Outcome
14.
J Immunother Cancer ; 9(10)2021 10.
Article in English | MEDLINE | ID: mdl-34599019

ABSTRACT

BACKGROUND: Atypical teratoid/rhabdoid tumors (AT/RT) are highly aggressive CNS tumors of infancy and early childhood. Hallmark is the surprisingly simple genome with inactivating mutations or deletions in the SMARCB1 gene as the oncogenic driver. Nevertheless, AT/RTs are infiltrated by immune cells and even clonally expanded T cells. However, it is unclear which epitopes T cells might recognize on AT/RT cells. METHODS: Here, we report a comprehensive mass spectrometry (MS)-based analysis of naturally presented human leukocyte antigen (HLA) class I and class II ligands on 23 AT/RTs. MS data were validated by matching with a human proteome dataset and exclusion of peptides that are part of the human benignome. Cryptic peptide ligands were identified using Peptide-PRISM. RESULTS: Comparative HLA ligandome analysis of the HLA ligandome revealed 55 class I and 139 class II tumor-exclusive peptides. No peptide originated from the SMARCB1 region. In addition, 61 HLA class I tumor-exclusive peptide sequences derived from non-canonically translated proteins. Combination of peptides from natural and cryptic class I and class II origin gave optimal representation of tumor cell compartments. Substantial overlap existed with the cryptic immunopeptidome of glioblastomas, but no concordance was found with extracranial tumors. More than 80% of AT/RT exclusive peptides were able to successfully prime CD8+ T cells, whereas naturally occurring memory responses in AT/RT patients could only be detected for class II epitopes. Interestingly, >50% of AT/RT exclusive class II ligands were also recognized by T cells from glioblastoma patients but not from healthy donors. CONCLUSIONS: These findings highlight that AT/RTs, potentially paradigmatic for other pediatric tumors with a low mutational load, present a variety of highly immunogenic HLA class I and class II peptides from canonical as well as non-canonical protein sources. Inclusion of such cryptic peptides into therapeutic vaccines would enable an optimized mapping of the tumor cell surface, thereby reducing the likelihood of immune evasion.


Subject(s)
Central Nervous System Neoplasms/immunology , Peptides/immunology , Rhabdoid Tumor/immunology , Adolescent , Central Nervous System Neoplasms/genetics , Central Nervous System Neoplasms/metabolism , Central Nervous System Neoplasms/therapy , Child , Child, Preschool , Female , HLA Antigens/genetics , HLA Antigens/immunology , HLA Antigens/metabolism , Humans , Immunohistochemistry , Immunotherapy , Male , Mass Spectrometry , Oncogenes , Peptides/metabolism , Peptides, Cyclic , Rhabdoid Tumor/genetics , Rhabdoid Tumor/metabolism , Rhabdoid Tumor/therapy
15.
Front Immunol ; 12: 689435, 2021.
Article in English | MEDLINE | ID: mdl-34671342

ABSTRACT

The complement system is a highly conserved component of innate immunity that is involved in recognizing and responding to pathogens. The system serves as a bridge between innate and adaptive immunity, and modulation of the complement system can affect the entire host immune response to a foreign insult. Neoplastic diseases have been shown to engage the complement system in order to evade the immune system, gain a selective growth advantage, and co-opt the surrounding environment for tumor proliferation. Historically, the central nervous system has been considered to be an immune-privileged environment, but it is now clear that there are active roles for both innate and adaptive immunity within the central nervous system. Much of the research on the role of immunological modulation of neoplastic disease within the central nervous system has focused on adaptive immunity, even though innate immunity still plays a critical role in the natural history of central nervous system neoplasms. Here, we review the modulation of the complement system by a variety of neoplastic diseases of the central nervous system. We also discuss gaps in the current body of knowledge and comment on future directions for investigation.


Subject(s)
Central Nervous System Neoplasms/metabolism , Central Nervous System/metabolism , Complement System Proteins/metabolism , Neuroimmunomodulation , Adaptive Immunity , Animals , Central Nervous System/immunology , Central Nervous System/pathology , Central Nervous System Neoplasms/immunology , Central Nervous System Neoplasms/pathology , Central Nervous System Neoplasms/therapy , Humans , Immunity, Innate , Prognosis , Signal Transduction
16.
Sci Rep ; 11(1): 20839, 2021 10 21.
Article in English | MEDLINE | ID: mdl-34675316

ABSTRACT

Recent studies have shown that ANXA2 is important in the development of many cancers, while its role in glioma-related immune response remains unclear. We aimed to comprehensively investigate its biological characteristics and clinical value in glioma. We analyzed 699 glioma samples from The Cancer Genome Atlas as training cohort and 325 samples from the Chinese Glioma Genome Atlas as validation cohort. All the statistical analyses and figures were generated with R. ANXA2 was overexpressed significantly in high-grade glioma, isocitrate dehydrogenase wild-type and mesenchymal-subtype glioma. ANXA2 was a special indicator of mesenchymal subtype. The survival analysis showed that highly-expressed ANXA2 was related to worse survival status as an independent factor of poor prognosis. Further gene ontology analysis showed that ANXA2 was mainly involved in immune response and inflammatory activities of glioma. Subsequent correlation analysis showed that ANXA2 was positively correlated with HCK, LCK, MHC II, STAT1 and interferon but negatively with IgG. Meanwhile, ANXA2 was positively related to the infiltration of tumor-related macrophages, regulatory T cells and myeloid-derived suppressor cells. Our study revealed that ANXA2 is a biomarker closely related to the malignant phenotype and poor prognosis of glioma, and plays an important role in immune response, inflammatory activity and immunosuppression.


Subject(s)
Annexin A2/genetics , Central Nervous System Neoplasms/genetics , Glioma/genetics , Up-Regulation , Annexin A2/immunology , Central Nervous System Neoplasms/diagnosis , Central Nervous System Neoplasms/immunology , Gene Expression Regulation, Neoplastic , Glioma/diagnosis , Glioma/immunology , Humans , Immune Tolerance , Prognosis
17.
Lancet Haematol ; 8(10): e711-e722, 2021 Oct.
Article in English | MEDLINE | ID: mdl-34560014

ABSTRACT

BACKGROUND: CNS relapse of acute lymphocytic leukaemia is difficult to treat. Durable remissions of relapsed or refractory B-cell acute lymphocytic leukaemia have been observed following treatment with CD19-directed chimeric antigen receptor (CAR) T cells; however, most trials have excluded patients with active CNS disease. We aimed to assess the safety and activity of CAR T-cell therapy in patients with a history of CNS relapsed or refractory B-cell acute lymphocytic leukaemia. METHODS: In this post-hoc analysis, we included 195 patients (aged 1-29 years; 110 [56%] male and 85 [44%] female) with relapsed or refractory CD19-positive acute lymphocytic leukaemia or lymphocytic lymphoma from five clinical trials (Pedi CART19, 13BT022, ENSIGN, ELIANA, and 16CT022) done at the Children's Hospital of Philadelphia (Philadelphia, PA, USA), in which participants received CD19-directed CAR T-cell therapy between April 17, 2012, and April 16, 2019. The trials required control of CNS disease at enrolment and infusion and excluded treatment in the setting of acute neurological toxic effects (>grade 1 in severity) or parenchymal lesions deemed to increase the risk of neurotoxicity. 154 patients from Pedi CART19, ELIANA, ENSIGN, and 16CT022 received tisagenlecleucel and 41 patients from the 13BT022 trial received the humanised CD19-directed CAR, huCART19. We categorised patients into two strata on the basis of CNS status at relapse or within the 12 months preceding CAR T-cell infusion-either CNS-positive or CNS-negative disease. Patients with CNS-positive disease were further divided on the basis of morphological bone marrow involvement-either combined bone marrow and CNS involvement, or isolated CNS involvement. Endpoints were the proportion of patients with complete response at 28 days after infusion, Kaplan-Meier analysis of relapse-free survival and overall survival, and the incidence of cytokine release syndrome and neurotoxicity. FINDINGS: Of all 195 patients, 66 (34%) were categorised as having CNS-positive disease and 129 (66%) as having CNS-negative disease, and 43 (22%) were categorised as having isolated CNS involvement. The median length of follow-up was 39 months (IQR 25-49) in the CNS-positive stratum and 36 months (18-49) in the CNS-negative stratum. The proportion of patients in the CNS-positive stratum with a complete response at 28 days after infusion was similar to that in the CNS-negative stratum (64 [97%] of 66 vs 121 [94%] of 129; p=0·74), with no significant difference in relapse-free survival (60% [95% CI 49-74] vs 60% [51-71]; p=0·50) or overall survival (83% [75-93] vs 71% [64-79]; p=0·39) at 2 years between the two groups. Overall survival at 2 years was significantly higher in patients with isolated CNS involvement compared with those with bone marrow involvement (91% [82-100] vs 71% [64-78]; p=0·046). The incidence and severity of neurotoxicity (any grade, 53 [41%] vs 38 [58%]; grade 1, 24 [19%] vs 20 [30%]; grade 2, 14 [11%] vs 10 [15%]; grade 3, 12 [9%] vs 6 [9%], and grade 4, 3 [2%] vs 2 [3%]; p=0·20) and cytokine release syndrome (any grade, 110 [85%] vs 53 [80%]; grade 1, 12 [9%] vs 2 [3%]; grade 2, 61 [47%] vs 38 [58%]; grade 3, 18 [14%] vs 7 [11%] and grade 4, 19 [15%] vs 6 [9%]; p=0·26) did not differ between the CNS-negative and the CNS-positive disease strata. INTERPRETATION: Tisagenlecleucel and huCART19 are active at clearing CNS disease and maintaining durable remissions in children and young adults with CNS relapsed or refractory B-cell acute lymphocytic leukaemia or lymphocytic lymphoma, without increasing the risk of severe neurotoxicity; although care should be taken in the timing of therapy and disease control to mitigate this risk. These preliminary findings support the use of these CAR T-cell therapies for patients with CNS relapsed or refractory B-cell acute lymphocytic leukaemia. FUNDING: Children's Hospital of Philadelphia Frontier Program.


Subject(s)
Antigens, CD19/immunology , Central Nervous System Neoplasms/therapy , Immunotherapy, Adoptive , Precursor Cell Lymphoblastic Leukemia-Lymphoma/therapy , Adolescent , Adult , Central Nervous System Neoplasms/immunology , Child, Preschool , Female , Humans , Infant , Male , Precursor Cell Lymphoblastic Leukemia-Lymphoma/immunology , Receptors, Chimeric Antigen/immunology , Recurrence , Young Adult
18.
Hematol Oncol ; 39(5): 625-638, 2021 Dec.
Article in English | MEDLINE | ID: mdl-34543472

ABSTRACT

Primary central nervous system lymphoma (PCNSL) is an aggressive and rare malignancy with poor prognosis. However, there are no reliable prognostic biomarkers for PCNSL in clinical practice. Here, we aimed to identify a reliable prognostic biomarker for predicting the survival of PCNSL patients. In this study, multiplex immunofluorescence and digital imaging analysis were used to characterize tumor-associated macrophages (TAMs) immunophenotypes and the expression of programmed cell death ligand 1 on TAMs, with regard to prognosis from diagnostic tumor tissue samples of 59 PCNSL patients. We found that the M2-to-M1 ratio was a more reliable prognostic biomarker for PCNSL than M1-like or M2-like macrophage infiltration. In addition, the combination of programmed death-ligand 1 (PD-L1) expression on TAMs and the M2-to-M1 ratio in PCNSL demonstrated improved performance in prognostic discrimination than PD-L1-positive TAMs or M2-to-M1 ratio. To validate the prognostic significance of the combined TAMs associated biomarkers, they were integrated into the International Extranodal Lymphoma Study Group (IELSG) index and termed as IELSG-M index. Kaplan-Meier plots showed that the IELSG-M index could discriminate patients into low-, intermediate- or high-risk subgroups, better than IELSG, in terms of prognosis. The areas under the receiver operating characteristic curves of IELSG-M was 0.844 for overall survival; superior to the IELSG model (0.580). Taken together, this study's findings showed that the combination of PD-L1 on TAMs and the M2-to-M1 ratio could be strong prognostic predictive biomarkers for PCNSL and the IELSG-M index had improved prognostic significance than the IELSG index.


Subject(s)
B7-H1 Antigen/metabolism , Biomarkers, Tumor/analysis , Central Nervous System Neoplasms/mortality , Lymphoma/mortality , Tumor Microenvironment , Tumor-Associated Macrophages/immunology , Central Nervous System Neoplasms/immunology , Central Nervous System Neoplasms/metabolism , Central Nervous System Neoplasms/pathology , Female , Follow-Up Studies , Humans , Lymphoma/immunology , Lymphoma/metabolism , Lymphoma/pathology , Male , Middle Aged , Prognosis , Retrospective Studies , Survival Rate
19.
Front Immunol ; 12: 693200, 2021.
Article in English | MEDLINE | ID: mdl-34290712

ABSTRACT

Background: The efficacy and safety of chimeric antigen receptor T (CAR-T) cell therapy in the treatment of non-Hodgkin's lymphoma has already been demonstrated. However, patients with a history of/active secondary central nervous system (CNS) lymphoma were excluded from the licensing trials conducted on two widely used CAR-T cell products, Axicabtagene ciloleucel (Axi-cel) and Tisagenlecleucel (Tisa-cel). Hence, the objective of the present review was to assess whether secondary CNS lymphoma patients would derive a benefit from Axi-cel or Tisa-cel therapy, while maintaining controllable safety. Method: Two reviewers searched PubMed, Embase, Web of Science, and Cochrane library independently in order to identify all records associated with Axi-cel and Tisa-cel published prior to February 15, 2021. Studies that included secondary CNS lymphoma patients treated with Axi-cel and Tisa-cel and reported or could be inferred efficacy and safety endpoints of secondary CNS lymphoma patients were included. A tool designed specifically to evaluate the risk of bias in case series and reports and the ROBINS-I tool applied for cohort studies were used. Results: Ten studies involving forty-four patients were included. Of these, seven were case reports or series. The other three reports were cohort studies involving twenty-five patients. Current evidence indicates that secondary CNS lymphoma patients could achieve long-term remission following Axi-cel and Tisa-cel treatment. Compared with the non-CNS cohort, however, progression-free survival and overall survival tended to be shorter. This was possibly due to the relatively small size of the CNS cohort. The incidence and grades of adverse effects in secondary CNS lymphoma patients resembled those in the non-CNS cohort. No incidences of CAR-T cell-related deaths were reported. Nevertheless, the small sample size introduced a high risk of bias and prevented the identification of specific patients who could benefit more from CAR-T cell therapy. Conclusion: Secondary CNS lymphoma patients could seem to benefit from both Axi-cel and Tisa-cel treatment, with controllable risks. Thus, CAR-T cell therapy has potential as a candidate treatment for lymphoma patients with CNS involvement. Further prospective studies with larger samples and longer follow-up periods are warranted and recommended.


Subject(s)
Biological Products/therapeutic use , Central Nervous System Neoplasms/therapy , Immunotherapy, Adoptive , Lymphoma/therapy , Receptors, Antigen, T-Cell/genetics , T-Lymphocytes/transplantation , Adolescent , Adult , Aged , Aged, 80 and over , Biological Products/adverse effects , Central Nervous System Neoplasms/immunology , Central Nervous System Neoplasms/mortality , Female , Humans , Immunotherapy, Adoptive/adverse effects , Lymphoma/immunology , Lymphoma/mortality , Male , Middle Aged , Patient Safety , Risk Assessment , Risk Factors , T-Lymphocytes/immunology , Treatment Outcome , Young Adult
20.
Blood Cancer J ; 11(7): 131, 2021 07 15.
Article in English | MEDLINE | ID: mdl-34267187

ABSTRACT

Chimeric antigen receptor (CAR) T-cell immunotherapy following autologous stem cell transplantation (ASCT) is a promising method for refractory or relapsed multiple myeloma, but explicit data for central nervous system lymphoma (CNSL) are lacking. Here, we treated 13 CNSL patients with ASCT sequential CD19/22 CAR T-cell infusion and simultaneously evaluated the clinical efficacy and toxicity. The 13 CNSL patients analyzed included four primary CNSL and nine secondary CNSL patients. Patients 1 and 10, who had complete remission status before enrollment, maintained clinical efficacy without recurrence. Nine of the remaining 11 patients responded to our protocol with a median durable time of 14.03 months, and the overall response and complete remission rate were 81.81% and 54.55%, respectively. No patient suffered grades 3-4 cytokine-release syndrome (CRS), and only patient 10 experienced severe immune effector cell-associated neurotoxicity syndrome (ICANS). In addition, increases in serum ferritin and interleukin-6 levels were often accompanied by CRS and ICANS. After a median follow-up time of 14.20 months, the estimated 1-year progression-free survival and overall survival rates were 74.59% and 82.50%, respectively. Sequential CD19/22 CAR T-cell immunotherapy following ASCT as a novel method for CNSL appears to have encouraging long-term efficacy with relatively manageable side effects.


Subject(s)
Central Nervous System Neoplasms/therapy , Hematopoietic Stem Cell Transplantation , Immunotherapy, Adoptive/methods , Lymphoma/therapy , Adult , Antigens, CD19/immunology , Antigens, CD20/immunology , Central Nervous System Neoplasms/immunology , Female , Humans , Lymphoma/immunology , Male , Middle Aged , Receptors, Chimeric Antigen/immunology , Receptors, Chimeric Antigen/therapeutic use , Transplantation, Autologous , Young Adult
SELECTION OF CITATIONS
SEARCH DETAIL