Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 242
Filter
1.
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
2.
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
3.
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
4.
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
5.
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
6.
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
7.
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
8.
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
9.
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
10.
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
12.
Front Immunol ; 12: 634031, 2021.
Article in English | MEDLINE | ID: mdl-34163465

ABSTRACT

Although there are several immunotherapy approaches for the treatment of Central Nervous System (CNS) tumors under evaluation, currently none of these approaches have received approval from the regulatory agencies. CNS tumors, especially glioblastomas, are tumors characterized by highly immunosuppressive tumor microenvironment, limiting the possibility of effectively eliciting an immune response. Moreover, the peculiar anatomic location of these tumors poses relevant challenges in terms of safety, since uncontrolled hyper inflammation could lead to cerebral edema and cranial hypertension. The most promising strategies of immunotherapy in neuro-oncology consist of the use of autologous T cells redirected against tumor cells through chimeric antigen receptor (CAR) constructs or genetically modified T-cell receptors. Trials based on native or genetically engineered oncolytic viruses and on vaccination with tumor-associated antigen peptides are also under evaluation. Despite some sporadic complete remissions achieved in clinical trials, the outcome of patients with CNS tumors treated with different immunotherapeutic approaches remains poor. Based on the lessons learned from these unsatisfactory experiences, novel immune-therapy approaches aimed at overcoming the profound immunosuppressive microenvironment of these diseases are bringing new hope to reach the cure for CNS tumors.


Subject(s)
Central Nervous System Neoplasms/therapy , Immunotherapy/trends , Medical Oncology/trends , Oncolytic Virotherapy/trends , Animals , Cancer Vaccines/therapeutic use , Central Nervous System Neoplasms/immunology , Central Nervous System Neoplasms/pathology , Diffusion of Innovation , Humans , Immune Checkpoint Inhibitors/therapeutic use , Immunotherapy, Adoptive/trends , Receptors, Chimeric Antigen/genetics , Receptors, Chimeric Antigen/immunology , T-Lymphocytes/immunology , T-Lymphocytes/transplantation , Treatment Outcome , Tumor Microenvironment
13.
Front Immunol ; 12: 600659, 2021.
Article in English | MEDLINE | ID: mdl-33833751

ABSTRACT

Adoptive T cell therapy has emerged as a revolutionary immunotherapy for treating cancer. Despite immense promise and clinical success in some hematologic malignancies, limitations remain that thwart its efficacy in solid tumors. Particularly in tumors of the central nervous system (CNS), T cell therapy is often restricted by the difficulty in intratumoral delivery across anatomical niches, suboptimal T cell specificity or activation, and intratumoral T cell dysfunction due to immunosuppressive tumor microenvironments (TMEs). Nanoparticles may offer several advantages to overcome these limitations of T cell therapy, as they can be designed to robustly and specifically activate T cells ex vivo prior to adoptive transfer, to encapsulate T cell stimulating agents for co-localized stimulation, and to be conjugated onto T cells for added functionality. This perspective highlights recent preclinical advances in using nanoparticles to enhance T cell therapy, and discusses the potential applicability and constraints of nanoparticle-enhanced T cells as a new platform for treating CNS tumors.


Subject(s)
Central Nervous System Neoplasms , Immunotherapy, Adoptive , Nanoparticles/therapeutic use , T-Lymphocytes/immunology , Tumor Microenvironment , Central Nervous System Neoplasms/immunology , Central Nervous System Neoplasms/therapy , Humans , Tumor Microenvironment/drug effects , Tumor Microenvironment/immunology
14.
Cancer Lett ; 509: 121-129, 2021 07 01.
Article in English | MEDLINE | ID: mdl-33766752

ABSTRACT

Primary central nervous system lymphoma (PCNSL) is an aggressive cancer typically confined to the brain, eyes, leptomeninges and spinal cord, without evidence of systemic involvement. PCNSL remains a challenge for scientists and clinicians due to insufficient biological knowledge, a lack of appropriate animal models and validated diagnostic biomarkers. We summarize recent findings on genomic, transcriptomic and epigenetic alterations identified in PCNSL. These findings help to define pathobiology of the disease and delineate defects in B cell differentiation. Evidence from genomic and transcriptomic studies helps to separate PCNSL from other hematological malignancies, improves diagnostics and reveals new therapeutic targets for treatment. Discovery of the CNS lymphatic system may be instrumental in better understanding the origin of the disease. We critically assess the attempts to model PCNSL in rodents, and conclude that there is a lack of a genetic/transgenic model that adequately mimics pathogenesis of the disease. Contribution of the tumor microenvironment in tumorigenesis and aggressiveness of PCNSL remains understudied. Assessing heterogeneity of immune infiltrates, cytokine profiling and molecular markers, may improve diagnostics and put forward new therapeutic strategies.


Subject(s)
Central Nervous System Neoplasms/pathology , Lymphoma/pathology , Neoplastic Stem Cells/pathology , Tumor Microenvironment , Animals , Apoptosis , Cell Differentiation , Cell Proliferation , Central Nervous System Neoplasms/genetics , Central Nervous System Neoplasms/immunology , Central Nervous System Neoplasms/metabolism , Epigenesis, Genetic , Gene Expression Regulation, Neoplastic , Humans , Lymphoma/genetics , Lymphoma/immunology , Lymphoma/metabolism , Neoplastic Stem Cells/immunology , Neoplastic Stem Cells/metabolism , Phenotype , Signal Transduction
15.
Theranostics ; 11(8): 3565-3579, 2021.
Article in English | MEDLINE | ID: mdl-33664848

ABSTRACT

Rationale: Primary central nervous system diffuse large B-cell lymphoma (PCNSL) is a rare and aggressive entity that resides in an immune-privileged site. The tumor microenvironment (TME) and the disruption of the immune surveillance influence lymphoma pathogenesis and immunotherapy resistance. Despite growing knowledge on heterogeneous therapeutic responses, no comprehensive description of the PCNSL TME is available. We hence investigated the immune subtypes of PCNSL and their association with molecular signaling and survival. Methods: Analysis of PCNSL transcriptomes (sequencing, n = 20; microarrays, n = 34). Integrated correlation analysis and signaling pathway topology enabled us to infer intercellular interactions. Immunohistopathology and digital imaging were used to validate bioinformatic results. Results: Transcriptomics revealed three immune subtypes: immune-rich, poor, and intermediate. The immune-rich subtype was associated to better survival and characterized by hyper-activation of STAT3 signaling and inflammatory signaling, e.g., IFNγ and TNF-α, resembling the hot subtype described in primary testicular lymphoma and solid cancer. WNT/ß-catenin, HIPPO, and NOTCH signaling were hyper-activated in the immune-poor subtype. HLA down-modulation was clearly associated with a low or intermediate immune infiltration and the absence of T-cell activation. Moreover, HLA class I down-regulation was also correlated with worse survival with implications on immune-intermediate PCNSL that frequently feature reduced HLA expression. A ligand-receptor intercellular network revealed high expression of two immune checkpoints, i.e., CTLA-4/CD86 and TIM-3/LAGLS9. TIM-3 and galectin-9 proteins were clearly upregulated in PCNSL. Conclusion: Altogether, our study reveals that patient stratification according to immune subtypes, HLA status, and immune checkpoint molecule quantification should be considered prior to immune checkpoint inhibitor therapy. Moreover, TIM-3 protein should be considered an axis for future therapeutic development.


Subject(s)
Central Nervous System Neoplasms/immunology , Lymphoma, Large B-Cell, Diffuse/immunology , Adult , Aged , Aged, 80 and over , Central Nervous System Neoplasms/metabolism , Central Nervous System Neoplasms/pathology , Cohort Studies , Down-Regulation , Female , Gene Expression Profiling , HLA Antigens/genetics , Humans , Kaplan-Meier Estimate , Lymphoma, Large B-Cell, Diffuse/metabolism , Lymphoma, Large B-Cell, Diffuse/pathology , Male , Middle Aged , Precision Medicine , Prognosis , Signal Transduction/genetics , Signal Transduction/immunology , Tumor Microenvironment/genetics , Tumor Microenvironment/immunology
16.
Mol Diagn Ther ; 25(2): 207-229, 2021 03.
Article in English | MEDLINE | ID: mdl-33646562

ABSTRACT

Cerebrospinal fluid (CSF) is a clear and paucicellular fluid that circulates within the ventricular system and the subarachnoid space of the central nervous system (CNS), and diverse CNS disorders can impact its composition, volume, and flow. As conventional CSF testing suffers from suboptimal sensitivity, this review aimed to evaluate the role of next-generation sequencing (NGS) in the work-up of infectious, neoplastic, neuroimmunological, and neurodegenerative CNS diseases. Metagenomic NGS showed improved sensitivity-compared to traditional methods-to detect bacterial, viral, parasitic, and fungal infections, while the overall performance was maximized in some studies when all diagnostic modalities were used. In patients with primary CNS cancer, NGS findings in the CSF were largely concordant with the molecular signatures derived from tissue-based molecular analysis; of interest, additional mutations were identified in the CSF in some glioma studies, reflecting intratumoral heterogeneity. In patients with metastasis to the CNS, NGS facilitated diagnosis, prognosis, therapeutic management, and monitoring, exhibiting higher sensitivity than neuroimaging, cytology, and plasma-based molecular analysis. Although evidence is still rudimentary, NGS could enhance the diagnosis and pathogenetic understanding of multiple sclerosis in addition to Alzheimer and Parkinson disease. To conclude, NGS has shown potential to aid the research, facilitate the diagnostic approach, and improve the management outcomes of all the aforementioned CNS diseases. However, to establish its role in clinical practice, the clinical validity and utility of each NGS protocol should be determined. Lastly, as most evidence has been derived from small and retrospective studies, results from randomized control trials could be of significant value.


Subject(s)
Central Nervous System Diseases/genetics , Central Nervous System Neoplasms/genetics , Immune System Diseases/genetics , Neurodegenerative Diseases/genetics , Central Nervous System/pathology , Central Nervous System Diseases/immunology , Central Nervous System Diseases/pathology , Central Nervous System Neoplasms/immunology , Central Nervous System Neoplasms/pathology , Humans , Immune System Diseases/immunology , Immune System Diseases/pathology , Mutation/genetics , Neurodegenerative Diseases/immunology , Neurodegenerative Diseases/pathology
17.
Clin Lymphoma Myeloma Leuk ; 21(4): e410-e414, 2021 04.
Article in English | MEDLINE | ID: mdl-33526401

ABSTRACT

BACKGROUND: chimeric antigen receptor-modified T cell (CAR-T) therapy is an effective and promising treatment for refractory and multiply relapsed B-cell acute lymphoblastic leukemia (B-ALL). Because of its side effects and poor responses such as neurotoxicity and cytokine release syndrome, patients with central nervous system leukemia were excluded in most previous clinical trials of CAR-T treatment. PATIENTS AND METHODS: We enrolled 3 B-ALL patients with central nervous system leukemia relapse. They were infused with CD19-specific CAR-Ts, and their clinical responses were evaluated by bone marrow smear, flow cytometry, and cytogenetic alterations detected by quantitative PCR, interleukin-6, and the expansion and persistence of circulating CAR-Ts in peripheral blood and cerebrospinal fluid. RESULTS: After CAR-T infusion, 2 of the 3 patients experienced bone marrow minimal residual disease-negative complete remission, and all patients tested negative for residual leukemia cells in cerebrospinal fluid tested by flow cytometry. These 3 patients experienced grade 2 or 3 cytokine release syndrome, which resolved completely after symptomatic treatment. None experienced neurotoxicity or needed further intensive care. CONCLUSION: CAR-T infusion is a potentially effective treatment for relapsed/refractory B-ALL patients with central nervous system involvement.


Subject(s)
Central Nervous System Neoplasms/therapy , Immunotherapy, Adoptive/adverse effects , Neoplasm Recurrence, Local/therapy , Precursor B-Cell Lymphoblastic Leukemia-Lymphoma/therapy , Bone Marrow/pathology , Central Nervous System Neoplasms/diagnosis , Central Nervous System Neoplasms/immunology , Child , Child, Preschool , Female , Humans , Immunotherapy, Adoptive/methods , Male , Neoplasm Recurrence, Local/diagnosis , Neoplasm Recurrence, Local/immunology , Neoplasm, Residual , Precursor B-Cell Lymphoblastic Leukemia-Lymphoma/diagnosis , Precursor B-Cell Lymphoblastic Leukemia-Lymphoma/immunology , Receptors, Chimeric Antigen/immunology , Treatment Outcome
18.
Cancer Immunol Immunother ; 70(7): 1979-1993, 2021 Jul.
Article in English | MEDLINE | ID: mdl-33416942

ABSTRACT

INTRODUCTION: Although recent clinical trials have demonstrated the efficacy of CD19-directed chimeric antigen receptor (CAR) T-cell therapy for refractory or relapsed B acute lymphoblastic leukemia (r/r B-ALL), most trials exclude patients with high-burden CNS leukemia (CNSL) to avoid the risk of severe neurotoxicity. There were only sparse cases describing the effect of CAR T cells on low-burden CNSL, and the safety and effectiveness of CAR T cells in high-burden CNSL remains unknown. METHODS: Here, we retrospectively analyzed the results of CD19 CAR T-cell therapy in 12 pediatric patients that had low (Blasts < 20/µL in CSF) or high-burdens (Blasts or intracranial solid mass) of CNS B-ALL, that are enrolled in three clinical trials and one pilot study at Beijing Boren Hospital RESULTS: Eleven patients (91.7%) achieved complete remission (CR) on day 30, and one patient got CR on day 90 after infusion. Most patient experienced mild cytokine-release syndrome. However, of the five patients who retained > 5/µL blasts in CSF or a solid mass before CAR T-cell expansion, four developed severe (grade 3-4) neurotoxicity featured by persistent cerebral edema and seizure, and they fully recovered after intensive managements. Sustained remission was achieved in 9 of the 12 patients, resulted in a 6-month leukemia-free survival rate of 81.8% (95% CI 59.0-100). Only one patient has CNS relapse again. CONCLUSION: Our study demonstrates that CAR T cells are effective in clearing both low- and high-burden CNSL, but a high CNSL burden before CAR T-cell expansion may cause severe neurotoxicity requiring intense intervention.


Subject(s)
Antigens, CD19/immunology , Central Nervous System Neoplasms/drug therapy , Cytokine Release Syndrome/pathology , Immunotherapy, Adoptive/adverse effects , Neurotoxicity Syndromes/pathology , Precursor B-Cell Lymphoblastic Leukemia-Lymphoma/drug therapy , Adolescent , Central Nervous System Neoplasms/immunology , Central Nervous System Neoplasms/pathology , Child , Child, Preschool , Cytokine Release Syndrome/etiology , Female , Follow-Up Studies , Humans , Infant , Male , Neurotoxicity Syndromes/etiology , Pilot Projects , Precursor B-Cell Lymphoblastic Leukemia-Lymphoma/immunology , Precursor B-Cell Lymphoblastic Leukemia-Lymphoma/pathology , Prognosis , Retrospective Studies , Survival Rate
19.
Transfusion ; 61(3): 744-753, 2021 03.
Article in English | MEDLINE | ID: mdl-33314235

ABSTRACT

BACKGROUND: Pediatric hematology-oncology patients require frequent platelet transfusions to manage chemotherapy-induced thrombocytopenia, and allergic transfusion reactions (ATRs) are common. Risk for platelet-associated ATRs can result from recipient- or donor-specific factors. STUDY DESIGN AND METHODS: We report a rare case in which an individual platelet donor caused repeated ATRs in multiple recipients. This observation led us to conduct a retrospective study at a pediatric hematology-oncology center to identify donor- and recipient-associated risk factors for ATRs. RESULTS: Single-donor platelets from an individual donor precipitated ATRs in 78.6% (n = 11/14) of recipients and 66.7% (n = 12/18) of platelet transfusions. We found in a cohort of pediatric hematology-oncology patients that 12.6% of recipients and 1.0% of platelet transfusions were associated with ATRs. Recipients who were aged 4 to 18 years, male, and those with central nervous system or solid tumors and with a history of ATRs to platelets were more likely to experience ATRs. Donor-associated risk factors were not identified, and we did not implicate additional donors in our single-center cohort with a frequency of ATRs comparable to the index donor. Based on our findings, we developed a novel statistical model to identify recipients and donors prone to experiencing or mediating ATRs. CONCLUSIONS: Both donors and recipients contribute to ATRs. Identification of high-risk donors and recipients for further scrutiny and potential interventions can improve the safety of platelet transfusions.


Subject(s)
Hematologic Neoplasms/blood , Hematologic Neoplasms/immunology , Platelet Transfusion/adverse effects , Transfusion Reaction/etiology , Adolescent , Adult , Aged , Blood Donors , Central Nervous System Neoplasms/blood , Central Nervous System Neoplasms/immunology , Child , Child, Preschool , Cohort Studies , Female , Hospitals , Humans , Infant , Male , Middle Aged , Models, Statistical , Retrospective Studies , Risk Factors , Thrombocytopenia/complications , Young Adult
20.
Blood ; 137(11): 1468-1477, 2021 03 18.
Article in English | MEDLINE | ID: mdl-33202420

ABSTRACT

Primary central nervous system lymphoma (PCNSL) is confined to the brain, eyes, and cerebrospinal fluid without evidence of systemic spread. Rarely, PCNSL occurs in the context of immunosuppression (eg, posttransplant lymphoproliferative disorders or HIV [AIDS-related PCNSL]). These cases are poorly characterized, have dismal outcome, and are typically Epstein-Barr virus (EBV)-associated (ie, tissue-positive). We used targeted sequencing and digital multiplex gene expression to compare the genetic landscape and tumor microenvironment (TME) of 91 PCNSL tissues all with diffuse large B-cell lymphoma histology. Forty-seven were EBV tissue-negative: 45 EBV- HIV- PCNSL and 2 EBV- HIV+ PCNSL; and 44 were EBV tissue-positive: 23 EBV+ HIV+ PCNSL and 21 EBV+ HIV- PCNSL. As with prior studies, EBV- HIV- PCNSL had frequent MYD88, CD79B, and PIM1 mutations, and enrichment for the activated B-cell (ABC) cell-of-origin subtype. In contrast, these mutations were absent in all EBV tissue-positive cases and ABC frequency was low. Furthermore, copy number loss in HLA class I/II and antigen-presenting/processing genes were rarely observed, indicating retained antigen presentation. To counter this, EBV+ HIV- PCNSL had a tolerogenic TME with elevated macrophage and immune-checkpoint gene expression, whereas AIDS-related PCNSL had low CD4 gene counts. EBV-associated PCNSL in the immunosuppressed is immunobiologically distinct from EBV- HIV- PCNSL, and, despite expressing an immunogenic virus, retains the ability to present EBV antigens. Results provide a framework for targeted treatment.


Subject(s)
Central Nervous System Neoplasms/etiology , Central Nervous System Neoplasms/immunology , Epstein-Barr Virus Infections/complications , Epstein-Barr Virus Infections/immunology , Herpesvirus 4, Human/immunology , Lymphoma/virology , Adult , Aged , Aged, 80 and over , Central Nervous System Neoplasms/genetics , Central Nervous System Neoplasms/virology , Epstein-Barr Virus Infections/genetics , Epstein-Barr Virus Infections/virology , Female , Herpesvirus 4, Human/isolation & purification , Humans , Immune Tolerance , Lymphoma/etiology , Male , Middle Aged , Mutation , Transcriptome , Tumor Microenvironment
SELECTION OF CITATIONS
SEARCH DETAIL
...