Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 17 de 17
Filter
1.
Nat Med ; 29(11): 2844-2853, 2023 Nov.
Article in English | MEDLINE | ID: mdl-37872225

ABSTRACT

The oncofetal antigen Claudin 6 (CLDN6) is highly and specifically expressed in many solid tumors, and could be a promising treatment target. We report dose escalation results from the ongoing phase 1/2 BNT211-01 trial evaluating the safety and feasibility of chimeric antigen receptor (CAR) T cells targeting the CLDN6 with or without a CAR-T cell-amplifying RNA vaccine (CARVac) at two dose levels (DLs) in relapsed/refractory CLDN6-positive solid tumors. The primary endpoints were safety and tolerability, maximum tolerated dose and recommended phase 2 dose (RP2D). Secondary endpoints included objective response rate (ORR) and disease control rate. We observed manageable toxicity, with 10 out of 22 patients (46%) experiencing cytokine release syndrome including one grade 3 event and 1 out of 22 (5%) with grade 1 immune effector cell-associated neurotoxicity syndrome. Dose-limiting toxicities occurred in two patients at the higher DL, resolving without sequelae. CAR-T cell engraftment was robust, and the addition of CARVac was well tolerated. The unconfirmed ORR in 21 evaluable patients was 33% (7 of 21), including one complete response. The disease control rate was 67% (14 of 21), with stable disease in seven patients. Patients with germ cell tumors treated at the higher DL exhibited the highest response rate (ORR 57% (4 of 7)). The maximum tolerated dose and RP2D were not established as the trial has been amended to utilize an automated manufacturing process. A repeat of the dose escalation is ongoing and will identify a RP2D for pivotal trials. ClinicalTrials.gov Identifier: NCT04503278 .


Subject(s)
Neoplasms , Humans , Neoplasms/drug therapy , T-Lymphocytes
2.
Science ; 367(6476): 446-453, 2020 01 24.
Article in English | MEDLINE | ID: mdl-31896660

ABSTRACT

Chimeric antigen receptor (CAR)-T cells have shown efficacy in patients with B cell malignancies. Yet, their application for solid tumors has challenges that include limited cancer-specific targets and nonpersistence of adoptively transferred CAR-T cells. Here, we introduce the developmentally regulated tight junction protein claudin 6 (CLDN6) as a CAR target in solid tumors and a strategy to overcome inefficient CAR-T cell stimulation in vivo. We demonstrate that a nanoparticulate RNA vaccine, designed for body-wide delivery of the CAR antigen into lymphoid compartments, stimulates adoptively transferred CAR-T cells. Presentation of the natively folded target on resident antigen-presenting cells promotes cognate and selective expansion of CAR-T cells. Improved engraftment of CAR-T cells and regression of large tumors in difficult-to-treat mouse models was achieved at subtherapeutic CAR-T cell doses.


Subject(s)
Cancer Vaccines/therapeutic use , Claudins/antagonists & inhibitors , Immunotherapy, Adoptive/methods , Receptors, Chimeric Antigen/immunology , Animals , Claudins/immunology , Female , Humans , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , RNA/therapeutic use , T-Lymphocytes/immunology , T-Lymphocytes/transplantation , Vaccines, Synthetic/therapeutic use
3.
Curr Gene Ther ; 15(4): 416-27, 2015.
Article in English | MEDLINE | ID: mdl-25981636

ABSTRACT

We report on a series of sequential events leading to long-term survival and cure of pediatric X-linked chronic granulomatous disease (X-CGD) patients after gamma-retroviral gene therapy (GT) and rescue HSCT. Due to therapyrefractory life-threatening infections requiring hematopoietic stem cell transplantation (HSCT) but absence of HLAidentical donors, we treated 2 boys with X-CGD by GT. Following GT both children completely resolved invasive Aspergillus nidulans infections. However, one child developed dual insertional activation of ecotropic viral integration site 1 (EVI1) and signal transducer and activator of transcription 3 (STAT3) genes, leading to myelodysplastic syndrome (MDS) with monosomy 7. Despite resistance to mismatched allo-HSCT with standard myeloablative conditioning, secondary intensified rescue allo-HSCT resulted in 100 % donor chimerism and disappearance of MDS. The other child did not develop MDS despite expansion of a clone with a single insertion in the myelodysplasia syndrome 1 (MDS1) gene and was cured by early standard allo-HSCT. The slowly developing dominance of clones harboring integrations in MDS1-EVI1 may guide clinical intervention strategies, i.e. early rescue allo-HSCT, prior to malignant transformation. GT was essential for both children to survive and to clear therapy-refractory infections, and future GT with safer lentiviral self-inactivated (SIN) vectors may offer a therapeutic alternative for X-CGD patients suffering from life-threatening infections and lacking HLA-identical HSC donors.


Subject(s)
Genetic Therapy/methods , Granulomatous Disease, Chronic/therapy , Hematopoietic Stem Cell Transplantation/methods , Aspergillosis/therapy , Aspergillus nidulans/pathogenicity , Child , Chromosome Deletion , Chromosomes, Human, Pair 7 , DNA-Binding Proteins/genetics , Gammaretrovirus/genetics , Genetic Therapy/adverse effects , Humans , MDS1 and EVI1 Complex Locus Protein , Male , Membrane Glycoproteins/genetics , Myelodysplastic Syndromes/etiology , NADPH Oxidase 2 , NADPH Oxidases/genetics , Proto-Oncogenes/genetics , STAT3 Transcription Factor/genetics , Transcription Factors/genetics
4.
Mol Ther ; 23(2): 330-8, 2015 Feb.
Article in English | MEDLINE | ID: mdl-25373520

ABSTRACT

Natural killer (NK) cells are an important effector cell type for adoptive cancer immunotherapy. Similar to T cells, NK cells can be modified to express chimeric antigen receptors (CARs) to enhance antitumor activity, but experience with CAR-engineered NK cells and their clinical development is still limited. Here, we redirected continuously expanding and clinically usable established human NK-92 cells to the tumor-associated ErbB2 (HER2) antigen. Following GMP-compliant procedures, we generated a stable clonal cell line expressing a humanized CAR based on ErbB2-specific antibody FRP5 harboring CD28 and CD3ζ signaling domains (CAR 5.28.z). These NK-92/5.28.z cells efficiently lysed ErbB2-expressing tumor cells in vitro and exhibited serial target cell killing. Specific recognition of tumor cells and antitumor activity were retained in vivo, resulting in selective enrichment of NK-92/5.28.z cells in orthotopic breast carcinoma xenografts, and reduction of pulmonary metastasis in a renal cell carcinoma model, respectively. γ-irradiation as a potential safety measure for clinical application prevented NK cell replication, while antitumor activity was preserved. Our data demonstrate that it is feasible to engineer CAR-expressing NK cells as a clonal, molecularly and functionally well-defined and continuously expandable cell therapeutic agent, and suggest NK-92/5.28.z cells as a promising candidate for use in adoptive cancer immunotherapy.


Subject(s)
Gene Expression , Killer Cells, Natural/immunology , Killer Cells, Natural/metabolism , Receptor, ErbB-2/immunology , Receptors, Immunologic/genetics , Receptors, Immunologic/immunology , Recombinant Fusion Proteins/genetics , Animals , Breast Neoplasms/genetics , Breast Neoplasms/immunology , Breast Neoplasms/therapy , Cell Line, Transformed , Cell Line, Tumor , Clonal Evolution , Cytotoxicity, Immunologic , Disease Models, Animal , Epitopes, T-Lymphocyte/immunology , Female , Genetic Vectors/genetics , Humans , Immunophenotyping , Immunotherapy , Lentivirus/genetics , Lymphocyte Culture Test, Mixed , Neoplasms/genetics , Neoplasms/immunology , Neoplasms/pathology , Neoplasms/therapy , Phenotype , Transduction, Genetic , Xenograft Model Antitumor Assays
5.
Sci Transl Med ; 6(227): 227ra33, 2014 Mar 12.
Article in English | MEDLINE | ID: mdl-24622513

ABSTRACT

Wiskott-Aldrich syndrome (WAS) is characterized by microthrombocytopenia, immunodeficiency, autoimmunity, and susceptibility to malignancies. In our hematopoietic stem cell gene therapy (GT) trial using a γ-retroviral vector, 9 of 10 patients showed sustained engraftment and correction of WAS protein (WASP) expression in lymphoid and myeloid cells and platelets. GT resulted in partial or complete resolution of immunodeficiency, autoimmunity, and bleeding diathesis. Analysis of retroviral insertion sites revealed >140,000 unambiguous integration sites and a polyclonal pattern of hematopoiesis in all patients early after GT. Seven patients developed acute leukemia [one acute myeloid leukemia (AML), four T cell acute lymphoblastic leukemia (T-ALL), and two primary T-ALL with secondary AML associated with a dominant clone with vector integration at the LMO2 (six T-ALL), MDS1 (two AML), or MN1 (one AML) locus]. Cytogenetic analysis revealed additional genetic alterations such as chromosomal translocations. This study shows that hematopoietic stem cell GT for WAS is feasible and effective, but the use of γ-retroviral vectors is associated with a substantial risk of leukemogenesis.


Subject(s)
Genetic Therapy/adverse effects , Mutagens/adverse effects , Wiskott-Aldrich Syndrome Protein/genetics , Wiskott-Aldrich Syndrome Protein/therapeutic use , Wiskott-Aldrich Syndrome/therapy , Adolescent , Animals , Blood Platelets/metabolism , Child , Child, Preschool , Clone Cells , Colitis/etiology , Disease Progression , Hematopoietic Stem Cell Transplantation , Hematopoietic Stem Cells/metabolism , Humans , Leukocytes, Mononuclear/metabolism , Lymphocytes/metabolism , Mice , Mice, Inbred NOD , Precursor T-Cell Lymphoblastic Leukemia-Lymphoma/etiology , Precursor T-Cell Lymphoblastic Leukemia-Lymphoma/pathology , Thrombocytopenia/therapy , Transplantation, Autologous , Treatment Outcome , Wiskott-Aldrich Syndrome/pathology , Wiskott-Aldrich Syndrome Protein/metabolism
6.
Cancer Immunol Immunother ; 63(2): 133-45, 2014 Feb.
Article in English | MEDLINE | ID: mdl-24190544

ABSTRACT

Adoptive cell therapy employing gene-modified T-cells expressing chimeric antigen receptors (CARs) has shown promising preclinical activity in a range of model systems and is now being tested in the clinical setting. The manufacture of CAR T-cells requires compliance with national and European regulations for the production of medicinal products. We established such a compliant process to produce T-cells armed with a first-generation CAR specific for carcinoembryonic antigen (CEA). CAR T-cells were successfully generated for 14 patients with advanced CEA(+) malignancy. Of note, in the majority of patients, the defined procedure generated predominantly CD4(+) CAR T-cells with the general T-cell population bearing an effector-memory phenotype and high in vitro effector function. Thus, improving the process to generate less-differentiated T-cells would be more desirable in the future for effective adoptive gene-modified T-cell therapy. However, these results confirm that CAR T-cells can be generated in a manner compliant with regulations governing medicinal products in the European Union.


Subject(s)
Adoptive Transfer , Carcinoembryonic Antigen/immunology , Chimerin Proteins/biosynthesis , Receptors, Antigen, T-Cell/biosynthesis , T-Lymphocytes/immunology , Humans , Immunophenotyping , Interferon-gamma/biosynthesis
7.
Hum Gene Ther Clin Dev ; 24(2): 86-98, 2013 Jun.
Article in English | MEDLINE | ID: mdl-23845071

ABSTRACT

Chronic granulomatous disease (CGD) is a primary immunodeficiency characterized by impaired antimicrobial activity in phagocytic cells. As a monogenic disease affecting the hematopoietic system, CGD is amenable to gene therapy. Indeed in a phase I/II clinical trial, we demonstrated a transient resolution of bacterial and fungal infections. However, the therapeutic benefit was compromised by the occurrence of clonal dominance and malignant transformation demanding alternative vectors with equal efficacy but safety-improved features. In this work we have developed and tested a self-inactivating (SIN) gammaretroviral vector (SINfes.gp91s) containing a codon-optimized transgene (gp91(phox)) under the transcriptional control of a myeloid promoter for the gene therapy of the X-linked form of CGD (X-CGD). Gene-corrected cells protected X-CGD mice from Aspergillus fumigatus challenge at low vector copy numbers. Moreover, the SINfes.gp91s vector generates substantial amounts of superoxide in human cells transplanted into immunodeficient mice. In vitro genotoxicity assays and longitudinal high-throughput integration site analysis in transplanted mice comprising primary and secondary animals for 11 months revealed a safe integration site profile with no signs of clonal dominance.


Subject(s)
Gammaretrovirus/genetics , Genetic Vectors/metabolism , Granulomatous Disease, Chronic/therapy , Animals , Aspergillus fumigatus/pathogenicity , Cells, Cultured , DNA Methylation , Disease Models, Animal , Drug Evaluation, Preclinical , Genetic Therapy , Genetic Vectors/genetics , Humans , Lung Diseases/microbiology , Lung Diseases/pathology , Membrane Glycoproteins/genetics , Membrane Glycoproteins/metabolism , Mice , NADPH Oxidase 2 , NADPH Oxidases/genetics , NADPH Oxidases/metabolism , Phenotype , Promoter Regions, Genetic , Proto-Oncogene Proteins c-fes/genetics , Superoxides/metabolism
8.
N Engl J Med ; 363(20): 1918-27, 2010 Nov 11.
Article in English | MEDLINE | ID: mdl-21067383

ABSTRACT

The Wiskott-Aldrich syndrome (WAS) is an X-linked recessive primary immunodeficiency disorder associated with thrombocytopenia, eczema, and autoimmunity. We treated two patients who had this disorder with a transfusion of autologous, genetically modified hematopoietic stem cells (HSC). We found sustained expression of WAS protein expression in HSC, lymphoid and myeloid cells, and platelets after gene therapy. T and B cells, natural killer (NK) cells, and monocytes were functionally corrected. After treatment, the patients' clinical condition markedly improved, with resolution of hemorrhagic diathesis, eczema, autoimmunity, and predisposition to severe infection. Comprehensive insertion-site analysis showed vector integration that targeted multiple genes controlling growth and immunologic responses in a persistently polyclonal hematopoiesis. (Funded by Deutsche Forschungsgemeinschaft and others; German Clinical Trials Register number, DRKS00000330.).


Subject(s)
Genetic Therapy , Hematopoietic Stem Cell Transplantation , Wiskott-Aldrich Syndrome Protein Family/genetics , Wiskott-Aldrich Syndrome/therapy , Gene Transfer Techniques , Genetic Therapy/adverse effects , Hematopoietic Stem Cell Transplantation/adverse effects , Humans , Infant , Male , Mutagenesis, Insertional , Transplantation, Autologous , Wiskott-Aldrich Syndrome/genetics , Wiskott-Aldrich Syndrome/immunology
10.
Nat Med ; 16(2): 198-204, 2010 Feb.
Article in English | MEDLINE | ID: mdl-20098431

ABSTRACT

Gene-modified autologous hematopoietic stem cells (HSC) can provide ample clinical benefits to subjects suffering from X-linked chronic granulomatous disease (X-CGD), a rare inherited immunodeficiency characterized by recurrent, often life-threatening bacterial and fungal infections. Here we report on the molecular and cellular events observed in two young adults with X-CGD treated by gene therapy in 2004. After the initial resolution of bacterial and fungal infections, both subjects showed silencing of transgene expression due to methylation of the viral promoter, and myelodysplasia with monosomy 7 as a result of insertional activation of ecotropic viral integration site 1 (EVI1). One subject died from overwhelming sepsis 27 months after gene therapy, whereas a second subject underwent an allogeneic HSC transplantation. Our data show that forced overexpression of EVI1 in human cells disrupts normal centrosome duplication, linking EVI1 activation to the development of genomic instability, monosomy 7 and clonal progression toward myelodysplasia.


Subject(s)
Chromosomes, Human, Pair 7 , DNA-Binding Proteins/genetics , Genetic Therapy , Genomic Instability , Granulomatous Disease, Chronic/therapy , Monosomy , Myelodysplastic Syndromes/genetics , Proto-Oncogenes/genetics , Transcription Factors/genetics , Adult , Humans , MDS1 and EVI1 Complex Locus Protein , NADPH Oxidases/metabolism , Promoter Regions, Genetic
11.
Nat Med ; 12(4): 401-9, 2006 Apr.
Article in English | MEDLINE | ID: mdl-16582916

ABSTRACT

Gene transfer into hematopoietic stem cells has been used successfully for correcting lymphoid but not myeloid immunodeficiencies. Here we report on two adults who received gene therapy after nonmyeloablative bone marrow conditioning for the treatment of X-linked chronic granulomatous disease (X-CGD), a primary immunodeficiency caused by a defect in the oxidative antimicrobial activity of phagocytes resulting from mutations in gp91(phox). We detected substantial gene transfer in both individuals' neutrophils that lead to a large number of functionally corrected phagocytes and notable clinical improvement. Large-scale retroviral integration site-distribution analysis showed activating insertions in MDS1-EVI1, PRDM16 or SETBP1 that had influenced regulation of long-term hematopoiesis by expanding gene-corrected myelopoiesis three- to four-fold in both individuals. Although insertional influences have probably reinforced the therapeutic efficacy in this trial, our results suggest that gene therapy in combination with bone marrow conditioning can be successfully used to treat inherited diseases affecting the myeloid compartment such as CGD.


Subject(s)
Carrier Proteins/genetics , DNA-Binding Proteins/genetics , Genetic Therapy/methods , Granulomatous Disease, Chronic/therapy , Hematopoietic Stem Cells/physiology , Neoplasm Proteins/genetics , Nuclear Proteins/genetics , Transcription Factors/genetics , Adult , Chromosomes, Human, X , Clinical Trials as Topic , Gene Transfer Techniques , Genetic Linkage , Genetic Markers , Genetic Vectors , Granulomatous Disease, Chronic/blood , Granulomatous Disease, Chronic/etiology , Granulomatous Disease, Chronic/genetics , Humans , MDS1 and EVI1 Complex Locus Protein , Mutagenesis, Insertional , Neutrophils/physiology , Proto-Oncogenes , RNA, Messenger/analysis , Retroviridae/genetics , Treatment Outcome
13.
Blood ; 102(12): 3934-7, 2003 Dec 01.
Article in English | MEDLINE | ID: mdl-12881303

ABSTRACT

Retroviral vectors are commonly used in clinical gene therapy, but recent observations of insertional oncogene activation in preclinical and clinical settings have forced a discussion of their safety. Here we investigated the relationship between retroviral transduction efficiency in mass cultures and the actual number of integrated vector copies in single cells using K562 leukemia and primary CD34+ cells. We found an exponential increase of integration numbers correlated to gene transfer rates and a linear increase of expression levels with insertion frequency. On average we detected one vector insertion per transduced cell for a gene transfer of less than 30%, 3 for 60%, and approximately 9 for 90% (in K562). Clonal analysis revealed strikingly increased variations of both transgene copy numbers (more than 20-fold in primary cells) and expression levels associated with higher transduction. Therefore, limiting retroviral gene transfer to approximately 30% may be suggested to avoid generating clones containing multiple insertions.


Subject(s)
Gene Dosage , Genetic Vectors/administration & dosage , Retroviridae/genetics , Transduction, Genetic/standards , Cell Culture Techniques , Clone Cells , Flow Cytometry , Gene Expression , Green Fluorescent Proteins , Hematopoietic Stem Cells/metabolism , Humans , K562 Cells , Luminescent Proteins/analysis , Luminescent Proteins/genetics
15.
Mol Ther ; 5(4): 473-8, 2002 Apr.
Article in English | MEDLINE | ID: mdl-11945075

ABSTRACT

Efficient retroviral gene transfer into primary cells is a prerequisite for various gene therapeutic strategies. We have developed a transduction protocol based on the preloading of tissue culture vessels with retroviral particles by low-speed (1000g) centrifugation. We show that vector-preloaded tissue culture vessels allow highly efficient gene transfer into various target cells. We obtained transduction rates of up to 85% for primary T lymphocytes after just a single round of transduction. Under clinically relevant conditions using a vector developed for suicide gene therapy and produced under good manufacturing practice (GMP) conditions, the described method allowed generation of large numbers (>2x10(9)) of gene-modified T cells. The preloading concept ensures transduction of target cells in their optimal growth medium regardless of the medium used for vector production. This facilitated highly efficient gene transfer into quite different target cells such as CD34(+) and AC133(+) bone marrow progenitor as well as mesenchymal stem cells. The presented method combines high gene-transfer rates with a great potential for standardization in accordance with GMP guidelines and is consequently well suited for both research and clinical applications. (c)2002 Elsevier Science (USA).


Subject(s)
Cell Culture Techniques/methods , Centrifugation , Genetic Vectors , Retroviridae/genetics , T-Lymphocytes/metabolism , Transduction, Genetic/methods , Cell Culture Techniques/instrumentation , Cell Line , Humans , Kinetics , Stem Cells/metabolism
16.
Int J Cancer ; 98(5): 785-92, 2002 Apr 10.
Article in English | MEDLINE | ID: mdl-11920652

ABSTRACT

This study was undertaken to analyze the hematotoxicity of paclitaxel (Taxol) and to test whether transduction of repopulating hematopoietic cells with a retroviral vector (SF1m) expressing the human multidrug resistance 1 gene (MDR1) would permit dose intensification following bone marrow transplantation (BMT). While the regimen chosen (8 x 20 mg/kg i.p. within 12 days) produced a non-lethal, reversible hematotoxicity in mice with steady-state hematopoiesis, only 35.3% (6/17) of control mice survived when treated starting 14 days post BMT. In contrast, 83.3% (15/18) of mice transplanted with SF1m-transduced cells survived, owing to a significant protection against severe acute myelotoxicity (as determined by neutrophil counts, white and red blood cell counts and values for hemoglobin and hematocrit). After recovery from chemotherapy, an increase of myeloid cells that were resistant to colchicine and effluxed the fluorochrome Rhodamine 123 was observed in SF1m-mice, but not in controls. These results reveal that the lethal, dose-limiting hematotoxicity of an intensified post-transplantation chemotherapy with paclitaxel can be prevented by retroviral transfer of the MDR1 gene to a minor proportion of repopulating cells. Our mouse model, mimicking clinically achievable gene transfer rates, thus suggests that bone marrow chemoprotection may widen the therapeutic window and permit an earlier onset of post-transplantation chemotherapy.


Subject(s)
Antineoplastic Agents, Phytogenic/administration & dosage , Genes, MDR/physiology , Hematopoietic Stem Cell Transplantation , Hematopoietic Stem Cells/drug effects , Paclitaxel/administration & dosage , Animals , Blood Cell Count , Colony-Forming Units Assay , Gene Transfer Techniques , Genetic Therapy/methods , Genetic Vectors , Hematopoietic Stem Cells/physiology , Humans , Mice , Mice, Inbred C57BL , Retroviridae/genetics
17.
Hum Gene Ther ; 13(2): 233-42, 2002 Jan 20.
Article in English | MEDLINE | ID: mdl-11812280

ABSTRACT

Myelosuppression is the main side effect of cancer chemotherapy. An improved rate of retroviral vector-mediated gene transfer to hematopoietic stem cells, shown in more recent clinical trials, has created the basis to test the concept of myeloprotective gene therapy. We transplanted clinical-scale human peripheral blood progenitor cell grafts (n = 2) transduced with retroviral vector SF91m3, which contains the human multidrug resistance 1 gene (MDR1), into nonobese diabetic/severe combined immunodeficient (NOD/SCID) mice. Engrafted mice of one cohort were protected from paclitaxel toxicity (p < 0.05) and we noted a similar trend in the second cohort. In paclitaxel-treated mice that had received gene-transduced cells we found a significant increase in gene marking (p < 0.05 - p < 0.01) or P-glycoprotein expression (p < 0.01) compared with their chemotherapy-naive counterparts. This is the first report showing that cytostatic drug resistance gene therapy can mediate chemoprotection of human clinically relevant stem cell populations with marrow engraftment potential.


Subject(s)
Genes, MDR/genetics , Hematopoietic Stem Cell Transplantation , Paclitaxel/toxicity , Animals , Animals, Genetically Modified , Drug Resistance/genetics , Gene Transfer Techniques , Genetic Therapy , Humans , Mice , Mice, SCID , Tumor Cells, Cultured
SELECTION OF CITATIONS
SEARCH DETAIL