Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 36
Filter
1.
Blood ; 124(8): 1277-87, 2014 Aug 21.
Article in English | MEDLINE | ID: mdl-24970931

ABSTRACT

A compact marker/suicide gene that utilizes established clinical-grade reagents and pharmaceuticals would be of considerable practical utility to T-cell cancer gene therapy. Marker genes enable measurement of transduction and allow selection of transduced cells, whereas suicide genes allow selective deletion of administered T cells in the face of toxicity. We have created a highly compact marker/suicide gene for T cells combining target epitopes from both CD34 and CD20 antigens (RQR8). This construct allows selection with the clinically approved CliniMACS CD34 system (Miltenyi). Further, the construct binds the widely used pharmaceutical antibody rituximab, resulting in selective deletion of transgene-expressing cells. We have tested the functionality of RQR8 in vitro and in vivo as well as in combination with T-cell engineering components. We predict that RQR8 will make T-cell gene therapy both safer and cheaper.


Subject(s)
Antigens, CD20/immunology , Antigens, CD34/immunology , Genes, Transgenic, Suicide , Genetic Therapy/methods , Neoplasms/therapy , T-Lymphocytes/transplantation , Allografts , Animals , Antigens, CD20/genetics , Antigens, CD20/metabolism , Antigens, CD34/genetics , Antigens, CD34/metabolism , Epitopes , Mice , Mice, Inbred BALB C , Neoplasms/genetics , Neoplasms/immunology , Neoplasms/metabolism , T-Lymphocytes/immunology , T-Lymphocytes/metabolism
2.
N Engl J Med ; 365(18): 1673-83, 2011 Nov 03.
Article in English | MEDLINE | ID: mdl-22047558

ABSTRACT

BACKGROUND: Cellular therapies could play a role in cancer treatment and regenerative medicine if it were possible to quickly eliminate the infused cells in case of adverse events. We devised an inducible T-cell safety switch that is based on the fusion of human caspase 9 to a modified human FK-binding protein, allowing conditional dimerization. When exposed to a synthetic dimerizing drug, the inducible caspase 9 (iCasp9) becomes activated and leads to the rapid death of cells expressing this construct. METHODS: We tested the activity of our safety switch by introducing the gene into donor T cells given to enhance immune reconstitution in recipients of haploidentical stem-cell transplants. Patients received AP1903, an otherwise bioinert small-molecule dimerizing drug, if graft-versus-host disease (GVHD) developed. We measured the effects of AP1903 on GVHD and on the function and persistence of the cells containing the iCasp9 safety switch. RESULTS: Five patients between the ages of 3 and 17 years who had undergone stem-cell transplantation for relapsed acute leukemia were treated with the genetically modified T cells. The cells were detected in peripheral blood from all five patients and increased in number over time, despite their constitutive transgene expression. A single dose of dimerizing drug, given to four patients in whom GVHD developed, eliminated more than 90% of the modified T cells within 30 minutes after administration and ended the GVHD without recurrence. CONCLUSIONS: The iCasp9 cell-suicide system may increase the safety of cellular therapies and expand their clinical applications. (Funded by the National Heart, Lung, and Blood Institute and the National Cancer Institute; ClinicalTrials.gov number, NCT00710892.).


Subject(s)
Caspase 9/genetics , Genes, Transgenic, Suicide , Graft vs Host Disease/therapy , Immunotherapy, Adoptive , T-Lymphocytes/transplantation , Tacrolimus Binding Proteins/genetics , Adolescent , Apoptosis , Caspase 9/metabolism , Child , Child, Preschool , Female , Gene Transfer Techniques , Humans , Leukemia/therapy , Male , Organic Chemicals/therapeutic use , Recurrence , Stem Cell Transplantation , T-Lymphocytes/immunology
3.
Genome Med ; 16(1): 8, 2024 01 09.
Article in English | MEDLINE | ID: mdl-38195504

ABSTRACT

BACKGROUND: As normal cells transform into cancers, their cell state changes, which may drive cancer cells into a stem-like or more primordial, foetal, or embryonic cell state. The transcriptomic profile of this final state may encode information about cancer's origin and how cancers relate to their normal cell counterparts. METHODS: Here, we used single-cell atlases to study cancer transformation in transcriptional terms. We utilised bulk transcriptomes across a wide spectrum of adult and childhood cancers, using a previously established method to interrogate their relationship to normal cell states. We extend and validate these findings using single-cell cancer transcriptomes and organ-specific atlases of colorectal and liver cancer. RESULTS: Our bulk transcriptomic data reveals that adult cancers rarely return to an embryonic state, but that a foetal state is a near-universal feature of childhood cancers. This finding was confirmed with single-cell cancer transcriptomes. CONCLUSIONS: Our findings provide a nuanced picture of transformation in human cancer, indicating cancer-specific rather than universal patterns of transformation pervade adult epithelial cancers.


Subject(s)
Liver Neoplasms , Adult , Humans , Liver Neoplasms/genetics , Embryonic Development , Fetus , Gene Expression Profiling , Transcriptome
4.
Cancer Cell ; 42(2): 283-300.e8, 2024 02 12.
Article in English | MEDLINE | ID: mdl-38181797

ABSTRACT

Pediatric patients with high-risk neuroblastoma have poor survival rates and urgently need more effective treatment options with less side effects. Since novel and improved immunotherapies may fill this need, we dissect the immunoregulatory interactions in neuroblastoma by single-cell RNA-sequencing of 24 tumors (10 pre- and 14 post-chemotherapy, including 5 pairs) to identify strategies for optimizing immunotherapy efficacy. Neuroblastomas are infiltrated by natural killer (NK), T and B cells, and immunosuppressive myeloid populations. NK cells show reduced cytotoxicity and T cells have a dysfunctional profile. Interaction analysis reveals a vast immunoregulatory network and identifies NECTIN2-TIGIT as a crucial immune checkpoint. Combined blockade of TIGIT and PD-L1 significantly reduces neuroblastoma growth, with complete responses (CR) in vivo. Moreover, addition of TIGIT+PD-L1 blockade to standard relapse treatment in a chemotherapy-resistant Th-ALKF1174L/MYCN 129/SvJ syngeneic model induces CR. In conclusion, our integrative analysis provides promising targets and a rationale for immunotherapeutic combination strategies.


Subject(s)
B7-H1 Antigen , Neuroblastoma , Humans , Child , Neoplasm Recurrence, Local , Neuroblastoma/drug therapy , Neuroblastoma/genetics , Receptors, Immunologic/genetics , Immunotherapy , Sequence Analysis, RNA
5.
Cancer Discov ; 14(4): 663-668, 2024 Apr 04.
Article in English | MEDLINE | ID: mdl-38571421

ABSTRACT

SUMMARY: We are building the world's first Virtual Child-a computer model of normal and cancerous human development at the level of each individual cell. The Virtual Child will "develop cancer" that we will subject to unlimited virtual clinical trials that pinpoint, predict, and prioritize potential new treatments, bringing forward the day when no child dies of cancer, giving each one the opportunity to lead a full and healthy life.


Subject(s)
Neoplasms , Humans , Neoplasms/genetics
6.
Front Oncol ; 13: 1167082, 2023.
Article in English | MEDLINE | ID: mdl-37091147

ABSTRACT

Clinical outcomes for many childhood brain tumours remain poor, despite our increasing understanding of the underlying disease biology. Advances in molecular diagnostics have refined our ability to classify tumour types and subtypes, and efforts are underway across multiple international paediatric neuro-oncology consortia to take novel biological insights in the worst prognosis entities into innovative clinical trials. Whilst for the first time we are designing such studies on the basis of disease-specific biological data, the levels of preclincial evidence in appropriate model systems on which these trials are initiated is still widely variable. We have considered these issues between CONNECT, PNOC and ITCC-Brain, and developed a framework in which we can assess novel concepts being brought forward for possible clinical translation. Whilst not intended to be proscriptive for every possible circumstance, these criteria provide a basis for self-assessment of evidence by laboratory scientists, and a platform for discussion and rational decision-making prior to moving forward clinically.

7.
Nat Med ; 29(4): 803-810, 2023 04.
Article in English | MEDLINE | ID: mdl-37024595

ABSTRACT

Cancer immunotherapies have unique toxicities. Establishment of grading scales and standardized grade-based treatment algorithms for toxicity syndromes can improve the safety of these treatments, as observed for cytokine release syndrome (CRS) and immune effector cell associated neurotoxicity syndrome (ICANS) in patients with B cell malignancies treated with chimeric antigen receptor (CAR) T cell therapy. We have observed a toxicity syndrome, distinct from CRS and ICANS, in patients treated with cell therapies for tumors in the central nervous system (CNS), which we term tumor inflammation-associated neurotoxicity (TIAN). Encompassing the concept of 'pseudoprogression,' but broader than inflammation-induced edema alone, TIAN is relevant not only to cellular therapies, but also to other immunotherapies for CNS tumors. To facilitate the safe administration of cell therapies for patients with CNS tumors, we define TIAN, propose a toxicity grading scale for TIAN syndrome and discuss the potential management of this entity, with the goal of standardizing both reporting and management.


Subject(s)
Neoplasms , Neurotoxicity Syndromes , Humans , Neoplasms/therapy , Immunotherapy, Adoptive/adverse effects , Immunotherapy , Inflammation , Cytokine Release Syndrome/etiology , Cytokine Release Syndrome/therapy , Neurotoxicity Syndromes/etiology
8.
Cancer Cell ; 40(1): 11-13, 2022 01 10.
Article in English | MEDLINE | ID: mdl-35016025

ABSTRACT

Chimeric antigen receptors (CARs) allow redirection of T cells against any surface antigen. However, CARs require optimization to achieve activity against low-density antigens. Heitzeneder et al. perform an iterative adjustment of CAR components to reach a design for targeting cerebroglycan (GPC2) that shows potent pre-clinical activity in neuroblastoma models.


Subject(s)
Neuroblastoma , Receptors, Chimeric Antigen , Glypicans , Humans , Receptors, Chimeric Antigen/genetics , T-Lymphocytes
9.
Commun Biol ; 5(1): 884, 2022 09 07.
Article in English | MEDLINE | ID: mdl-36071103

ABSTRACT

A fundamental step of tumour single cell mRNA analysis is separating cancer and non-cancer cells. We show that the common approach to separation, using shifts in average expression, can lead to erroneous biological conclusions. By contrast, allelic imbalances representing copy number changes directly detect the cancer genotype and accurately separate cancer from non-cancer cells. Our findings provide a definitive approach to identifying cancer cells from single cell mRNA sequencing data.


Subject(s)
Neoplasms , Transcriptome , Allelic Imbalance/genetics , Genotype , Humans , Neoplasms/diagnosis , Neoplasms/genetics , Polymorphism, Single Nucleotide , RNA, Messenger/genetics
10.
Nat Med ; 28(4): 743-751, 2022 04.
Article in English | MEDLINE | ID: mdl-35288693

ABSTRACT

KMT2A-rearranged infant ALL is an aggressive childhood leukemia with poor prognosis. Here, we investigated the developmental state of KMT2A-rearranged infant B-cell acute lymphoblastic leukemia (B-ALL) using bulk messenger RNA (mRNA) meta-analysis and examination of single lymphoblast transcriptomes against a developing bone marrow reference. KMT2A-rearranged infant B-ALL was uniquely dominated by an early lymphocyte precursor (ELP) state, whereas less adverse NUTM1-rearranged infant ALL demonstrated signals of later developing B cells, in line with most other childhood B-ALLs. We compared infant lymphoblasts with ELP cells and revealed that the cancer harbored hybrid myeloid-lymphoid features, including nonphysiological antigen combinations potentially targetable to achieve cancer specificity. We validated surface coexpression of exemplar combinations by flow cytometry. Through analysis of shared mutations in separate leukemias from a child with infant KMT2A-rearranged B-ALL relapsing as AML, we established that KMT2A rearrangement occurred in very early development, before hematopoietic specification, emphasizing that cell of origin cannot be inferred from the transcriptional state.


Subject(s)
Precursor Cell Lymphoblastic Leukemia-Lymphoma , Transcriptome , Bone Marrow/metabolism , Child , Gene Rearrangement/genetics , Humans , Infant , Mutation/genetics , Myeloid-Lymphoid Leukemia Protein/genetics , Precursor Cell Lymphoblastic Leukemia-Lymphoma/genetics , Transcriptome/genetics
11.
Blood ; 114(23): 4792-803, 2009 Nov 26.
Article in English | MEDLINE | ID: mdl-19770360

ABSTRACT

Epstein-Barr virus (EBV)-driven posttransplantation lymphoproliferative disease (PTLD) is a serious complication of immunosuppression after either stem cell transplantation (SCT) or solid organ transplantation (SOT). Adoptive transfer of EBV-specific cytotoxic T lymphocytes (EBV-CTLs) is an effective prophylaxis and treatment for PTLD after SCT, but not for PTLD after SOT when pharmacologic immunosuppression cannot be discontinued. We report the generation of calcineurin (CN) mutants that render EBV-CTL resistant to the immunosuppressants tacrolimus (FK506) and cyclosporin A (CsA): mutant CNa12 confers resistance to CsA but not FK506, and mutant CNa22 confers resistance to FK506 but not CsA, whereas mutant CNb30 renders CTLs resistant to both calcineurin inhibitors. Untransduced EBV-CTLs do not proliferate in the presence of FK506/CsA. However, EBV-CTLs transduced with a retroviral vector coding for these mutants retain the ability to both proliferate and secrete normal levels of interferon-gamma in the presence therapeutic levels of FK506 (CNa12), CsA (CNa22), or both (CNb30). The cytotoxicity and phenotype of EBV-CTL lines were unaffected by expression of these mutant CNs. This approach should allow effective immunotherapy with EBV-CTLs in the SOT setting without risking the graft by reduction in immunosuppression, and represents a generic approach to improving immunotherapy in the face of immunosuppression.


Subject(s)
Calcineurin/genetics , Cyclosporine/pharmacology , Epstein-Barr Virus Infections/therapy , Genetic Vectors/pharmacology , Immunosuppressive Agents/pharmacology , Lymphoproliferative Disorders/therapy , Postoperative Complications/therapy , T-Lymphocytes, Cytotoxic/drug effects , Tacrolimus/pharmacology , Adoptive Transfer , Calcineurin/chemistry , Calcineurin Inhibitors , Cells, Cultured/drug effects , Cells, Cultured/immunology , Cyclophilin A/metabolism , Cytotoxicity, Immunologic , Drug Resistance/genetics , Humans , Interferon-gamma/metabolism , Jurkat Cells/drug effects , Jurkat Cells/immunology , Models, Molecular , Molecular Sequence Data , Mutagenesis, Site-Directed , Organ Transplantation , Protein Conformation , Protein Interaction Mapping , Recombinant Fusion Proteins/antagonists & inhibitors , Recombinant Fusion Proteins/genetics , Retroviridae/genetics , T-Lymphocytes, Cytotoxic/immunology , T-Lymphocytes, Cytotoxic/metabolism , T-Lymphocytes, Cytotoxic/transplantation , Tacrolimus Binding Protein 1A/chemistry , Tacrolimus Binding Protein 1A/genetics , Tacrolimus Binding Protein 1A/metabolism
12.
Blood ; 113(11): 2442-50, 2009 Mar 12.
Article in English | MEDLINE | ID: mdl-18971421

ABSTRACT

Treatment of Epstein-Barr virus (EBV)-positive nasopharyngeal carcinoma (NPC) with EBV-specific cytotoxic T cells (EBV-specific CTL) has been promising, producing clinical responses. However, infused EBV-specific CTL did not expand in vivo, likely limiting their antitumor activity. Lymphodepleting patients with chemotherapy before T-cell transfer enhances in vivo T-cell expansion, but results in nonspecific destruction of the resident immune system and can have significant toxicity. To evaluate if monoclonal antibodies (mAbs) can produce a more selective lymphodepletion, we conducted a clinical study in which NPC patients received a pair of lymphodepleting mAbs targeted to the CD45 antigen (CD45 mAbs) before EBV-specific CTL infusion. Eight patients with recurrent NPC received CD45 mAbs followed by escalating doses of autologous EBV-specific CTL. Infusion of CD45 mAbs resulted in transient lymphopenia in all patients and an increase in interleukin-15 (IL-15) levels in 6 out 8 patients. All patients had an increase in their peripheral blood frequency of EBV-specific T cells after CTL infusion. Three patients with a persistent increase had clinical benefits including 1 complete response (> 24 months) and 2 with stable disease (for 12 and 15 months). Lymphodepleting mAbs prior CTL transfer may represent an alternative to chemotherapy to enhance expansion of infused CTL. This study is registered at (http://www.clinialtrials.gov) as NCT00608257.


Subject(s)
Antibodies, Monoclonal/therapeutic use , Carcinoma/therapy , Herpesvirus 4, Human/immunology , Immunotherapy, Adoptive/methods , Leukocyte Common Antigens/immunology , Nasopharyngeal Neoplasms/therapy , T-Lymphocytes, Cytotoxic/transplantation , Adolescent , Adult , Antibodies, Monoclonal/pharmacology , Carcinoma/complications , Carcinoma/immunology , Epstein-Barr Virus Infections/complications , Female , Humans , Immunosuppressive Agents/pharmacology , Immunosuppressive Agents/therapeutic use , Male , Middle Aged , Nasopharyngeal Neoplasms/complications , Nasopharyngeal Neoplasms/immunology , T-Lymphocytes, Cytotoxic/immunology , Time Factors , Transplantation Conditioning/methods
13.
Nat Commun ; 12(1): 444, 2021 01 19.
Article in English | MEDLINE | ID: mdl-33469002

ABSTRACT

Glioblastoma multiforme (GBM) is the most common and aggressive form of primary brain cancer, for which effective therapies are urgently needed. Chimeric antigen receptor (CAR)-based immunotherapy represents a promising therapeutic approach, but it is often impeded by highly immunosuppressive tumor microenvironments (TME). Here, in an immunocompetent, orthotopic GBM mouse model, we show that CAR-T cells targeting tumor-specific epidermal growth factor receptor variant III (EGFRvIII) alone fail to control fully established tumors but, when combined with a single, locally delivered dose of IL-12, achieve durable anti-tumor responses. IL-12 not only boosts cytotoxicity of CAR-T cells, but also reshapes the TME, driving increased infiltration of proinflammatory CD4+ T cells, decreased numbers of regulatory T cells (Treg), and activation of the myeloid compartment. Importantly, the immunotherapy-enabling benefits of IL-12 are achieved with minimal systemic effects. Our findings thus show that local delivery of IL-12 may be an effective adjuvant for CAR-T cell therapy for GBM.


Subject(s)
Brain Neoplasms/therapy , Glioblastoma/therapy , Immunoconjugates/administration & dosage , Immunotherapy, Adoptive/methods , Interleukin-12/administration & dosage , Animals , Brain/diagnostic imaging , Brain/immunology , Brain/pathology , Brain Neoplasms/diagnostic imaging , Brain Neoplasms/immunology , Brain Neoplasms/pathology , Cell Line, Tumor/transplantation , Disease Models, Animal , ErbB Receptors/immunology , Female , Glioblastoma/diagnostic imaging , Glioblastoma/immunology , Glioblastoma/pathology , Humans , Immunoconjugates/immunology , Immunoglobulin Fc Fragments/administration & dosage , Immunoglobulin Fc Fragments/immunology , Injections, Intralesional/methods , Interleukin-12/immunology , Magnetic Resonance Imaging, Interventional , Mice , Receptors, Chimeric Antigen/immunology , Single-Chain Antibodies/administration & dosage , Single-Chain Antibodies/immunology , T-Lymphocytes, Regulatory/immunology , Tumor Microenvironment/immunology
14.
Nat Commun ; 12(1): 1407, 2021 03 03.
Article in English | MEDLINE | ID: mdl-33658498

ABSTRACT

Malignant rhabdoid tumour (MRT) is an often lethal childhood cancer that, like many paediatric tumours, is thought to arise from aberrant fetal development. The embryonic root and differentiation pathways underpinning MRT are not firmly established. Here, we study the origin of MRT by combining phylogenetic analyses and single-cell mRNA studies in patient-derived organoids. Comparison of somatic mutations shared between cancer and surrounding normal tissues places MRT in a lineage with neural crest-derived Schwann cells. Single-cell mRNA readouts of MRT differentiation, which we examine by reverting the genetic driver mutation underpinning MRT, SMARCB1 loss, suggest that cells are blocked en route to differentiating into mesenchyme. Quantitative transcriptional predictions indicate that combined HDAC and mTOR inhibition mimic MRT differentiation, which we confirm experimentally. Our study defines the developmental block of MRT and reveals potential differentiation therapies.


Subject(s)
Mutation , Rhabdoid Tumor/genetics , Rhabdoid Tumor/pathology , Cell Differentiation/genetics , DNA Methylation , Drug Screening Assays, Antitumor , Gene Expression Profiling , Gene Expression Regulation, Neoplastic/drug effects , Histone Deacetylase Inhibitors/pharmacology , Humans , Neural Crest/pathology , Phylogeny , Rhabdoid Tumor/drug therapy , SMARCB1 Protein/genetics , Single-Cell Analysis , TOR Serine-Threonine Kinases/antagonists & inhibitors , Tissue Culture Techniques/methods
15.
Sci Adv ; 7(6)2021 02.
Article in English | MEDLINE | ID: mdl-33547074

ABSTRACT

Neuroblastoma is a childhood cancer that resembles developmental stages of the neural crest. It is not established what developmental processes neuroblastoma cancer cells represent. Here, we sought to reveal the phenotype of neuroblastoma cancer cells by comparing cancer (n = 19,723) with normal fetal adrenal single-cell transcriptomes (n = 57,972). Our principal finding was that the neuroblastoma cancer cell resembled fetal sympathoblasts, but no other fetal adrenal cell type. The sympathoblastic state was a universal feature of neuroblastoma cells, transcending cell cluster diversity, individual patients, and clinical phenotypes. We substantiated our findings in 650 neuroblastoma bulk transcriptomes and by integrating canonical features of the neuroblastoma genome with transcriptional signals. Overall, our observations indicate that a pan-neuroblastoma cancer cell state exists, which may be attractive for novel immunotherapeutic and targeted avenues.


Subject(s)
Neural Stem Cells , Neuroblastoma , Child , Humans , Neural Crest/metabolism , Neural Stem Cells/metabolism , Neuroblastoma/genetics , Neuroblastoma/metabolism , Neuroblastoma/pathology , RNA, Messenger/genetics , Transcriptome
16.
Nat Commun ; 12(1): 3896, 2021 06 23.
Article in English | MEDLINE | ID: mdl-34162837

ABSTRACT

Tumor cells may share some patterns of gene expression with their cell of origin, providing clues into the differentiation state and origin of cancer. Here, we study the differentiation state and cellular origin of 1300 childhood and adult kidney tumors. Using single cell mRNA reference maps of normal tissues, we quantify reference "cellular signals" in each tumor. Quantifying global differentiation, we find that childhood tumors exhibit fetal cellular signals, replacing the presumption of "fetalness" with a quantitative measure of immaturity. By contrast, in adult cancers our assessment refutes the suggestion of dedifferentiation towards a fetal state in most cases. We find an intimate connection between developmental mesenchymal populations and childhood renal tumors. We demonstrate the diagnostic potential of our approach with a case study of a cryptic renal tumor. Our findings provide a cellular definition of human renal tumors through an approach that is broadly applicable to human cancer.


Subject(s)
Kidney Neoplasms/genetics , Kidney/metabolism , RNA, Messenger/genetics , RNA-Seq/methods , Single-Cell Analysis/methods , Transcriptome , Adult , Algorithms , Child , Fetus/metabolism , Gene Expression Regulation, Developmental , Humans , Kidney/embryology , Kidney Neoplasms/embryology , Kidney Neoplasms/metabolism , Models, Genetic , Signal Transduction/genetics
17.
J Exp Med ; 200(12): 1623-33, 2004 Dec 20.
Article in English | MEDLINE | ID: mdl-15611290

ABSTRACT

Epstein Barr virus (EBV)+ Hodgkin's disease (HD) expresses clearly identified tumor antigens derived from the virus and could, in principle, be a target for adoptive immunotherapy with viral antigen-specific T cells. However, like most tumor-associated antigens in immunocompetent hosts, these potential targets are only weakly immunogenic, consisting primarily of the latent membrane protein (LMP)1 and LMP2 antigens. Moreover, Hodgkin tumors possess a range of tumor evasion strategies. Therefore, the likely value of immunotherapy with EBV-specific cytotoxic effector cells has been questioned. We have now used a combination of gene marking, tetramer, and functional analyses to track the fate and assess the activity of EBV cytotoxic T lymphocyte (CTL) lines administered to 14 patients treated for relapsed EBV+ HD. Gene marking studies showed that infused effector cells could further expand by several logs in vivo, contribute to the memory pool (persisting up to 12 mo), and traffic to tumor sites. Tetramer and functional analyses showed that T cells reactive with the tumor-associated antigen LMP2 were present in the infused lines, expanded in peripheral blood after infusion, and also entered tumor. Viral load decreased, demonstrating the biologic activity of the infused CTLs. Clinically, EBV CTLs were well tolerated, could control type B symptoms (fever, night sweats, and weight loss), and had antitumor activity. After CTL infusion, five patients were in complete remission at up to 40 mo, two of whom had clearly measurable tumor at the time of treatment. One additional patient had a partial response, and five had stable disease. The performance and fate of these human tumor antigen-specific T cells in vivo suggests that they might be of value for the treatment of EBV+ Hodgkin lymphoma.


Subject(s)
Herpesviridae Infections/therapy , Herpesvirus 4, Human/immunology , Hodgkin Disease/therapy , Immunotherapy, Adoptive , T-Lymphocytes, Cytotoxic/transplantation , Adolescent , Adult , Cell Movement/immunology , Child , Female , Herpesviridae Infections/immunology , Herpesviridae Infections/pathology , Herpesviridae Infections/virology , Hodgkin Disease/immunology , Hodgkin Disease/pathology , Hodgkin Disease/virology , Humans , Male , Prognosis , Remission Induction , T-Lymphocytes, Cytotoxic/immunology , Tumor Escape/immunology , Viral Load , Viral Matrix Proteins/immunology
18.
Lancet ; 374(9693): 912-20, 2009 Sep 12.
Article in English | MEDLINE | ID: mdl-19729196

ABSTRACT

BACKGROUND: Stem-cell transplantation can cure primary immunodeficiencies. However, in patients with pre-existing organ toxicity, patients younger than 1 year, and those with DNA or telomere repair disorders, chemotherapy-based conditioning is poorly tolerated and results in major morbidity and mortality. We tested a novel antibody-based minimal-intensity conditioning (MIC) regimen to assess whether this approach allowed curative donor stem-cell engraftment without non-haemopoietic toxicity. METHODS: 16 high-risk patients underwent stem-cell transplantation for primary immunodeficiencies with an MIC regimen consisting of two rat anti-CD45 monoclonal antibodies YTH 24.5 and YTH 54.12 for myelosuppression, and alemtuzumab (anti-CD52) and fludarabine, and low dose cyclophosphamide for immunosuppression. Donors were matched siblings (n=5), and matched (9) and mismatched (2) unrelated donors. FINDINGS: Antibody-based conditioning was well tolerated, with only two cases of grade 3 and no grade 4 toxicity. Rates of clinically significant acute (n=6, 36%) and chronic graft-versus-host disease (GVHD) (n=5, 31%) were acceptable. 15 of 16 patients (94%) engrafted, of whom 11 (69%) achieved full or high-level mixed chimerism in both lymphoid and myeloid lineages, and three achieved engraftment in the T-lymphoid lineage only. One patient needed retransplantation. At a median of 40 months post-transplant, 13 of 16 patients (81%) in this high-risk cohort were alive and cured from their underlying disease. INTERPRETATION: Monoclonal antibody-based conditioning seems well tolerated and can achieve curative engraftment even in patients with severe organ toxicity or DNA repair defects, or both. This novel approach represents a shift from the paradigm that intensive chemotherapy or radiotherapy, or both, is needed for donor stem-cell engraftment. This antibody-based conditioning regimen may reduce toxicity and late effects and enable SCT in virtually any primary immunodeficiency patient with a matched donor. FUNDING: None.


Subject(s)
Antibodies, Monoclonal/therapeutic use , Hematopoietic Stem Cell Transplantation/methods , Immunologic Deficiency Syndromes/therapy , Immunologic Factors/therapeutic use , Leukocyte Common Antigens/antagonists & inhibitors , Transplantation Conditioning/methods , Alemtuzumab , Animals , Antibodies, Monoclonal, Humanized , Antibodies, Neoplasm/therapeutic use , Child, Preschool , Cyclophosphamide/therapeutic use , Disease-Free Survival , Female , Follow-Up Studies , Graft vs Host Disease/epidemiology , Graft vs Host Disease/etiology , Graft vs Host Disease/prevention & control , Hematopoietic Stem Cell Transplantation/adverse effects , Humans , Immunosuppressive Agents/therapeutic use , Infant , Kaplan-Meier Estimate , Male , Rats , Transplantation Chimera , Transplantation Conditioning/adverse effects , Transplantation Conditioning/mortality , Treatment Outcome , Vidarabine/analogs & derivatives , Vidarabine/therapeutic use
19.
Nat Commun ; 11(1): 4324, 2020 08 28.
Article in English | MEDLINE | ID: mdl-32859926

ABSTRACT

Immune-therapy is an attractive alternative therapeutic approach for targeting central nervous system (CNS) tumors and the constituency of the Tumor Immune Microenvironment (TIME) likely to predict patient response. Here, we describe the TIME of >6000 primarily pediatric CNS tumors using a deconvolution approach (methylCIBERSORT). We produce and validate a custom reference signature defining 11 non-cancer cell types to estimate relative proportions of infiltration in a panCNS tumor cohort spanning 80 subtypes. We group patients into three broad immune clusters associated with CNS tumor types/subtypes. In cohorts of medulloblastomas (n = 2325), malignant rhabdoid tumors (n = 229) and pediatric high-grade gliomas (n = 401), we show significant associations with molecular subgroups/subtypes, mutations, and prognosis. We further identify tumor-specific immune clusters with phenotypic characteristics relevant to immunotherapy response (i.e. Cytolytic score, PDL1 expression). Our analysis provides an indication of the potential future therapeutic and prognostic possibilities of immuno-methylomic profiling in pediatric CNS tumor patients that may ultimately inform approach to immune-therapy.


Subject(s)
Central Nervous System Neoplasms/immunology , Central Nervous System Neoplasms/therapy , Immunotherapy/methods , Tumor Microenvironment/immunology , Adolescent , Central Nervous System Neoplasms/genetics , Child , Child, Preschool , Cohort Studies , Glioma , Histones/genetics , Humans , Leukocytes , Medulloblastoma/immunology , Mutation , Prognosis , Rhabdoid Tumor
20.
Sci Transl Med ; 12(571)2020 11 25.
Article in English | MEDLINE | ID: mdl-33239386

ABSTRACT

The reprogramming of a patient's immune system through genetic modification of the T cell compartment with chimeric antigen receptors (CARs) has led to durable remissions in chemotherapy-refractory B cell cancers. Targeting of solid cancers by CAR-T cells is dependent on their infiltration and expansion within the tumor microenvironment, and thus far, fewer clinical responses have been reported. Here, we report a phase 1 study (NCT02761915) in which we treated 12 children with relapsed/refractory neuroblastoma with escalating doses of second-generation GD2-directed CAR-T cells and increasing intensity of preparative lymphodepletion. Overall, no patients had objective clinical response at the evaluation point +28 days after CAR-T cell infusion using standard radiological response criteria. However, of the six patients receiving ≥108/meter2 CAR-T cells after fludarabine/cyclophosphamide conditioning, two experienced grade 2 to 3 cytokine release syndrome, and three demonstrated regression of soft tissue and bone marrow disease. This clinical activity was achieved without on-target off-tumor toxicity. Targeting neuroblastoma with GD2 CAR-T cells appears to be a valid and safe strategy but requires further modification to promote CAR-T cell longevity.


Subject(s)
Neuroblastoma , Receptors, Chimeric Antigen , Child , Humans , Immunotherapy, Adoptive , Neoplasm Recurrence, Local , Neuroblastoma/therapy , Receptors, Antigen, T-Cell/genetics , Receptors, Chimeric Antigen/genetics , T-Lymphocytes , Tumor Microenvironment
SELECTION OF CITATIONS
SEARCH DETAIL