1
|
Ma J, Meyer S, Olweus J, Jin C, Yu D. An adjusted droplet digital PCR assay for quantification of vector copy number in CAR-T cell and TCR-T cell products. IMMUNO-ONCOLOGY TECHNOLOGY 2025; 25:101031. [PMID: 40236328 PMCID: PMC11997555 DOI: 10.1016/j.iotech.2024.101031] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Indexed: 04/17/2025]
Abstract
Background Genetically engineered T-cell therapy holds immense promise in cancer immunotherapy. These T-cell products are typically engineered by vectors that permanently integrate into the T-cell genome, thus raising concerns about potential risks of insertional mutagenesis. Therefore, it becomes imperative to assess the integrated vector copy number (VCN) as a critical safety parameter for gene-engineered cell products. Materials and methods In this study, we developed a robust assay for assessing the VCN of chimeric antigen receptor-T cell and T-cell receptor T-cell products, based on the droplet digital polymerase chain reaction (ddPCR) method. To provide accurate representation of the VCN in gene-engineered cells, we implemented a calculation that factors in the putative transduction efficiency based on Poisson distribution statistics. The adjusted VCN value (VCNadj) was also compared with VCN value from sorted transgene-positive cell populations, to validate its accuracy. Results This assay consistently and accurately determines the average VCN for cell products. By comparing the VCN in sorted transgene-positive cell populations, we validated the refinement calculation provides a closer approximation to the actual VCN within transduced cells, offering a more realistic representation of the VCN for engineered cell products. Conclusion In summary, we present a reliable and robust ddPCR-based assay for quantification of VCN in gene-engineered cell products.
Collapse
Affiliation(s)
- J. Ma
- Department of Immunology, Genetics and Pathology, Uppsala University, Uppsala, Sweden
- Elicera Therapeutics AB, Gothenburg, Sweden
| | - S. Meyer
- Department of Cancer Immunology, Institute for Cancer Research, Oslo University Hospital Radiumhospitalet, Oslo, Norway
- Precision Immunotherapy Alliance, University of Oslo, Oslo, Norway
| | - J. Olweus
- Department of Cancer Immunology, Institute for Cancer Research, Oslo University Hospital Radiumhospitalet, Oslo, Norway
- Precision Immunotherapy Alliance, University of Oslo, Oslo, Norway
| | - C. Jin
- Department of Immunology, Genetics and Pathology, Uppsala University, Uppsala, Sweden
- Elicera Therapeutics AB, Gothenburg, Sweden
| | - D. Yu
- Department of Immunology, Genetics and Pathology, Uppsala University, Uppsala, Sweden
- Elicera Therapeutics AB, Gothenburg, Sweden
| |
Collapse
|
2
|
Guo Z, He M, Liu N, Yang Y, Sun R, Wang J, Wang Q. In vitro functional validation of anti-CD19 chimeric antigen receptor T cells expressing lysine-specific demethylase 1 short hairpin RNA for the treatment of diffuse large B cell lymphoma. Front Immunol 2025; 15:1521778. [PMID: 39872520 PMCID: PMC11770093 DOI: 10.3389/fimmu.2024.1521778] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2024] [Accepted: 12/24/2024] [Indexed: 01/30/2025] Open
Abstract
Background Chimeric antigen receptor T (CAR-T) cell therapy is more effective in relapsed or refractory diffuse large B cell lymphoma (DLBCL) than other therapies, but a high proportion of patients relapse after CAR-T cell therapy owing to antigen escape, limited persistence of CAR-T cells, and immunosuppression in the tumor microenvironment. CAR-T cell exhaustion is a major cause of relapse. Epigenetic modifications can regulate T cell activation, maturation and depletion; they can be applied to reduce T cell depletion, improve infiltration, and promote memory phenotype formation to reduce relapse after CAR-T cell therapy. Purpose We propose to develop and validate in vitro the function of novel CAR-T cells for the treatment of DLBCL, which simultaneously express an anti-CD19 CAR with lysine-specific demethylase 1 (LSD1) short hairpin (sh)RNA to prevent depletion and prolong the survival of CAR-T cells. Methods We designed an shRNA sequence targeting LSD1 mRNA, and created a vector with the following elements: the U6 promoter driving expression of the LSD1 shRNA sequence, the EF1a promoter driving a second-generation anti-CD19 CAR sequence encoding an anti-CD19 single-chain variable fragment (FMC63), the CD8 hinge and transmembrane structural domains, the CD28 co-stimulatory structural domain, and the CD3ζ-activating structural domain. The MFG-LSD1 shRNA anti-CD19 CAR plasmid was first constructed, then packaged in retroviral vectors and transduced into human primary peripheral blood mononuclear cell-derived T cells to generate the corresponding CAR-T cells. We examined by flow cytometry the efficiency of two CAR-T cells in killing U-2932 cells (a human DLBCL line) upon co-culture with RNAU6 anti-CD19 CAR-T cells or LSD1 shRNA anti-CD19 CAR-T cells. We analyzed Ki-67 staining of the CAR-T cells by flow cytometry on days 0, 5, and 10, and counted the cells to assess expansion. We also used flow cytometry to detect the central memory T cell (TCM) proportion. Results We detected the expression of the CAR in the CAR-T cells by flow cytometry, and observed transduction rates of 31.5% for RNAU6 anti-CD19 CAR-T cells and 60.7% for LSD1 shRNA anti-CD19 CAR-T cells. The killing efficiency of LSD1 shRNA anti-CD19 CAR-T cells was significantly higher than that of RNAU6 anti-CD19 CAR-T cells at the low effector target ratio. We further found that LSD1 shRNA anti-CD19 CAR-T cells secreted more IFN-γ and granzyme B than RNAU6 anti-CD19 CAR-T cells. CAR-T cells proliferated after U-2932 cell stimulation and were able to sustain proliferation. After stimulation via U-2932 cell co-culture, both RNAU6 anti-CD19 CAR-T and LSD1 shRNA anti-CD19 CAR-T populations had increased proportions of cells with the TCM phenotype, with a higher percentage among LSD1 shRNA anti-CD19 CAR-T cells. Conclusion We developed a novel, feasible CD19-LSD1 shRNA CAR-T cell strategy for the treatment of DLBCL. Our in vitro assay results showed that LSD1 shRNA anti-CD19 CAR-T cells more effectively killed target cells than RNAU6 anti-CD19 CAR-T cells, and developed a higher proportion of TCM phenotype cells. LSD1 shRNA anti-CD19 CAR-T cells may represent a potential treatment for DLBCL.
Collapse
MESH Headings
- Humans
- Lymphoma, Large B-Cell, Diffuse/therapy
- Lymphoma, Large B-Cell, Diffuse/immunology
- Lymphoma, Large B-Cell, Diffuse/genetics
- Histone Demethylases/genetics
- Receptors, Chimeric Antigen/immunology
- Receptors, Chimeric Antigen/genetics
- Receptors, Chimeric Antigen/metabolism
- Antigens, CD19/immunology
- Immunotherapy, Adoptive/methods
- RNA, Small Interfering/genetics
- Cell Line, Tumor
- T-Lymphocytes/immunology
Collapse
Affiliation(s)
- Zhi Guo
- Institute of Infection, Immunology and Tumor Microenvironment, Hubei Province Key Laboratory of Occupational Hazard Identification and Control, School of Medicine, Wuhan University of Science and Technology, Wuhan, China
- Department of Hematology, The 6th Affiliated Hospital of Shenzhen University Health Science Center, Shenzhen, China
| | - Mingxin He
- Department of Hematology, The 6th Affiliated Hospital of Shenzhen University Health Science Center, Shenzhen, China
| | - Ning Liu
- Department of Hematology, The 6th Affiliated Hospital of Shenzhen University Health Science Center, Shenzhen, China
| | - Yiqing Yang
- Shenzhen Cell Valley Biomedical Co., LTD, Shenzhen, China
| | - Rui Sun
- Shenzhen Cell Valley Biomedical Co., LTD, Shenzhen, China
| | - Jianxun Wang
- Shenzhen Cell Valley Biomedical Co., LTD, Shenzhen, China
| | - Qiang Wang
- Institute of Infection, Immunology and Tumor Microenvironment, Hubei Province Key Laboratory of Occupational Hazard Identification and Control, School of Medicine, Wuhan University of Science and Technology, Wuhan, China
| |
Collapse
|
3
|
Xie Y, Li K, Yang L, Zeng X, Chen Z, Ma X, Zhang L, Zhou Y, Jin L, Yang Y, Lou X. Expanding the phenotypic and genetic spectrum of GTPBP3 deficiency: findings from nine Chinese pedigrees. Orphanet J Rare Dis 2024; 19:488. [PMID: 39719609 DOI: 10.1186/s13023-024-03469-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2024] [Accepted: 11/19/2024] [Indexed: 12/26/2024] Open
Abstract
BACKGROUND GTPBP3 catalyzes τm5(s2) U biosynthesis at the 34th wobble position of mitochondrial tRNAs, the hypomodification of τm5U leads to mitochondrial disease. While twenty-three variants of GTPBP3 have been reported worldwide, the genetic landscape in China remains uncertain. METHODS By using whole-exome sequencing, the candidate individuals carrying GTPBP3 variants were screened and identified. Pathogenicity analysis of variants was biochemically verified by patients-derived immortalized lymphocytes and cell models. RESULTS Through whole-exome sequencing, thirteen variants associated with GTPBP3 were identified in nine Chinese pedigrees, with eight of these variants being newly reported. Affected individuals displayed classic neurologic phenotypes and heart complications including developmental delay, seizures, hypotonia, exercise intolerance, and hypertrophic cardiomyopathy. Additionally, they displayed new symptoms such as eye problems like strabismus and heart issues related to valve function. Studies conducted on patient-derived cells provided evidence of reduced levels of GTPBP3 and impairment in mitochondrial energetic biogenesis. Re-expressing GTPBP3 variants in knockout cell lines further defined the pathogenicity of the novel variants. Analysis of the genetic spectrum in the Chinese population highlighted a concentration in exons 4 and 6, with c.689A > C being the prominent hotspot. CONCLUSION Our findings emphasize the extensive clinical and genetic implications of GTPBP3-related mitochondrial disorders, particularly within the Chinese population, but further investigations are needed to explore the phenotype-genotype correlation.
Collapse
Affiliation(s)
- Yaojun Xie
- Laboratory Medicine Center, Department of Genetic and Genomic Medicine, Zhejiang Provincial People's Hospital, Affiliated People's Hospital, Hangzhou Medical College, Hangzhou, Zhejiang, China
- Genetics Center of Obstetrics and Gynecology, Obstetrics and Gynecology Hospital of Fudan University, Shanghai, China
| | - Keyi Li
- Key Laboratory of Laboratory Medicine, Ministry of Education, Zhejiang Provincial Key Laboratory of Medical Genetics, School of Laboratory Medicine and Life Sciences, Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Li Yang
- Department of Pediatrics, Clinical Research Center for Children Neurodevelopmental Disabilities of Hunan Province, Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Xiaofei Zeng
- Key Laboratory of Laboratory Medicine, Ministry of Education, Zhejiang Provincial Key Laboratory of Medical Genetics, School of Laboratory Medicine and Life Sciences, Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Zhehui Chen
- Department of Pediatrics, Peking University First Hospital, Beijing, China
| | - Xue Ma
- Department of Pediatrics, Peking University First Hospital, Beijing, China
| | - Luyi Zhang
- Key Laboratory of Laboratory Medicine, Ministry of Education, Zhejiang Provincial Key Laboratory of Medical Genetics, School of Laboratory Medicine and Life Sciences, Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Yuwei Zhou
- Key Laboratory of Laboratory Medicine, Ministry of Education, Zhejiang Provincial Key Laboratory of Medical Genetics, School of Laboratory Medicine and Life Sciences, Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Liqin Jin
- Department of Scientific Research, Zhejiang Provincial People's Hospital, Affiliated People's Hospital, Hangzhou Medical College, Hangzhou, Zhejiang, China.
| | - Yanling Yang
- Department of Pediatrics, Peking University First Hospital, Beijing, China.
| | - Xiaoting Lou
- Laboratory Medicine Center, Department of Genetic and Genomic Medicine, Zhejiang Provincial People's Hospital, Affiliated People's Hospital, Hangzhou Medical College, Hangzhou, Zhejiang, China.
- Key Laboratory of Laboratory Medicine, Ministry of Education, Zhejiang Provincial Key Laboratory of Medical Genetics, School of Laboratory Medicine and Life Sciences, Wenzhou Medical University, Wenzhou, Zhejiang, China.
| |
Collapse
|
4
|
Derigs P, Schubert ML, Dreger P, Schmitt A, Yousefian S, Haas S, Röthemeier C, Neuber B, Hückelhoven-Krauss A, Brüggemann M, Bernhard H, Kobbe G, Lindemann A, Rummel M, Michels B, Korell F, Ho AD, Müller-Tidow C, Schmitt M. Third-generation anti-CD19 CAR T cells for relapsed/refractory chronic lymphocytic leukemia: a phase 1/2 study. Leukemia 2024; 38:2419-2428. [PMID: 39192036 PMCID: PMC11519001 DOI: 10.1038/s41375-024-02392-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2024] [Revised: 08/15/2024] [Accepted: 08/19/2024] [Indexed: 08/29/2024]
Abstract
Third-generation chimeric antigen receptor T cells (CARTs) for relapsed or refractory (r/r) chronic lymphocytic leukemia (CLL) may improve efficacy compared to second-generation CARTs due to their enhanced CAR design. We performed the first phase 1/2 investigator-initiated trial evaluating escalating doses of third-generation CARTs (HD-CAR-1) targeting CD19 in patients with r/r CLL and B-cell lymphoma. CLL eligibility criteria were failure to two therapy lines including at least one pathway inhibitor and/or allogeneic hematopoietic cell transplantation. Nine heavily pretreated patients received HD-CAR-1 at dose levels ranging from 1 × 106 to 200 × 106 CART/m2. In-house HD-CAR-1 manufacturing was successful for all patients. While neurotoxicity was absent, one case of grade 3 cytokine release syndrome was observed. By day 90, six patients (67%) attained a CR, five of these (83%) with undetectable MRD. With a median follow-up of 27 months, 2-year PFS and OS were 30% and 69%, respectively. HD-CAR-1 products of responders contained significantly more CD4 + T cells compared to non-responders. In non-responders, a strong enrichment of effector memory-like CD8 + T cells with high expression of CD39 and/or CD197 was observed. HD-CAR-1 demonstrated encouraging efficacy and exceptionally low treatment-specific toxicity, presenting new treatment options for patients with r/r CLL. Trial registration: #NCT03676504.
Collapse
MESH Headings
- Humans
- Leukemia, Lymphocytic, Chronic, B-Cell/therapy
- Leukemia, Lymphocytic, Chronic, B-Cell/immunology
- Male
- Immunotherapy, Adoptive/methods
- Immunotherapy, Adoptive/adverse effects
- Antigens, CD19/immunology
- Middle Aged
- Female
- Aged
- Receptors, Chimeric Antigen/immunology
- Neoplasm Recurrence, Local/pathology
- Neoplasm Recurrence, Local/immunology
- Neoplasm Recurrence, Local/therapy
- Adult
- Follow-Up Studies
Collapse
Affiliation(s)
- Patrick Derigs
- Internal Medicine V, Hematology, Oncology and Rheumatology, Heidelberg University Hospital, Heidelberg, Germany.
| | - Maria-Luisa Schubert
- Internal Medicine V, Hematology, Oncology and Rheumatology, Heidelberg University Hospital, Heidelberg, Germany
| | - Peter Dreger
- Internal Medicine V, Hematology, Oncology and Rheumatology, Heidelberg University Hospital, Heidelberg, Germany
| | - Anita Schmitt
- Internal Medicine V, Hematology, Oncology and Rheumatology, Heidelberg University Hospital, Heidelberg, Germany
| | - Schayan Yousefian
- Berlin Institute of Health (BIH) at Charité - Universitätsmedizin Berlin, Berlin, Germany
- Berlin Institute for Medical Systems Biology, Max Delbrück Center for Molecular Medicine in the Helmholtz Association, Berlin, Germany
- Department of Hematology, Oncology and Tumor Immunology, Charité University Medicine, Berlin, Germany
| | - Simon Haas
- Berlin Institute of Health (BIH) at Charité - Universitätsmedizin Berlin, Berlin, Germany
- Berlin Institute for Medical Systems Biology, Max Delbrück Center for Molecular Medicine in the Helmholtz Association, Berlin, Germany
- Department of Hematology, Oncology and Tumor Immunology, Charité University Medicine, Berlin, Germany
- German Cancer Consortium (DKTK) and German Cancer Research Center (DKFZ)/National Center for Tumor Diseases (NCT), Heidelberg, Germany
- Heidelberg Institute for Stem Cell Technology and Experimental Medicine (HI-STEM gGmbH), Heidelberg, Germany
- Precision Healthcare University Research Institute, Queen Mary University of London, London, UK
| | - Caroline Röthemeier
- Berlin Institute of Health (BIH) at Charité - Universitätsmedizin Berlin, Berlin, Germany
- Berlin Institute for Medical Systems Biology, Max Delbrück Center for Molecular Medicine in the Helmholtz Association, Berlin, Germany
- Department of Hematology, Oncology and Tumor Immunology, Charité University Medicine, Berlin, Germany
| | - Brigitte Neuber
- Internal Medicine V, Hematology, Oncology and Rheumatology, Heidelberg University Hospital, Heidelberg, Germany
| | - Angela Hückelhoven-Krauss
- Internal Medicine V, Hematology, Oncology and Rheumatology, Heidelberg University Hospital, Heidelberg, Germany
| | - Monika Brüggemann
- Department of Internal Medicine II, University Hospital Schleswig-Holstein, Kiel, Germany
| | - Helga Bernhard
- Department of Internal Medicine V, Klinikum Darmstadt, Darmstadt, Germany
| | - Guido Kobbe
- Department of Hematology, Oncology and Clinical Immunology, University Hospital Düsseldorf, Düsseldorf, Germany
| | | | - Mathias Rummel
- Department of Internal Medicine IV, University Hospital Giessen, Giessen, Germany
| | - Birgit Michels
- Internal Medicine V, Hematology, Oncology and Rheumatology, Heidelberg University Hospital, Heidelberg, Germany
| | - Felix Korell
- Internal Medicine V, Hematology, Oncology and Rheumatology, Heidelberg University Hospital, Heidelberg, Germany
| | - Anthony D Ho
- Internal Medicine V, Hematology, Oncology and Rheumatology, Heidelberg University Hospital, Heidelberg, Germany
- German Cancer Consortium (DKTK) and German Cancer Research Center (DKFZ)/National Center for Tumor Diseases (NCT), Heidelberg, Germany
| | - Carsten Müller-Tidow
- Internal Medicine V, Hematology, Oncology and Rheumatology, Heidelberg University Hospital, Heidelberg, Germany
- German Cancer Consortium (DKTK) and German Cancer Research Center (DKFZ)/National Center for Tumor Diseases (NCT), Heidelberg, Germany
| | - Michael Schmitt
- Internal Medicine V, Hematology, Oncology and Rheumatology, Heidelberg University Hospital, Heidelberg, Germany
- German Cancer Consortium (DKTK) and German Cancer Research Center (DKFZ)/National Center for Tumor Diseases (NCT), Heidelberg, Germany
| |
Collapse
|
5
|
Barisic S, Cherkasova E, Nadal R, Tian X, Chen L, Parrizzi A, Reger RN, Scurti GM, Nishimura MI, Childs RW. Quantification of circulating TCR-engineered T cells targeting a human endogenous retrovirus post-adoptive transfer using nanoplate digital PCR. Mol Ther Methods Clin Dev 2024; 32:101324. [PMID: 39319301 PMCID: PMC11419864 DOI: 10.1016/j.omtm.2024.101324] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2024] [Accepted: 08/15/2024] [Indexed: 09/26/2024]
Abstract
In vivo expansion of genetically modified T cells in cancer patients following adoptive transfer has been linked to both anti-tumor activity and T cell-mediated toxicities. The development of digital PCR has improved the accuracy in quantifying the in vivo status of adoptively infused T cells compared to qPCR or flow cytometry. Here, we developed and evaluated the feasibility and performance of nanoplate-based digital PCR (ndPCR) to quantify adoptively infused T cells engineered with a T cell receptor (TCR) that recognizes a human endogenous retrovirus type E (HERV-E) antigen. Analysis of blood samples collected from patients with metastatic kidney cancer following the infusion of HERV-E TCR-transduced T cells established the limit of detection of ndPCR to be 0.3 transgene copies/μL of reaction. The lower limit of quantification for ndPCR was one engineered T cell per 10,000 PBMCs, which outperformed both qPCR and flow cytometry by 1 log. High inter-test and test-retest reliability was confirmed by analyzing blood samples collected from multiple patients. In conclusion, we demonstrated the feasibility of ndPCR for detecting and monitoring the fate of TCR-engineered T cells in adoptive cell therapy.
Collapse
Affiliation(s)
- Stefan Barisic
- Laboratory of Transplantation Immunotherapy, Cellular and Molecular Therapeutics Branch, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD 20892, USA
| | - Elena Cherkasova
- Laboratory of Transplantation Immunotherapy, Cellular and Molecular Therapeutics Branch, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD 20892, USA
| | - Rosa Nadal
- Laboratory of Transplantation Immunotherapy, Cellular and Molecular Therapeutics Branch, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD 20892, USA
| | - Xin Tian
- Office of Biostatistics Research, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD 20892, USA
| | - Long Chen
- Laboratory of Transplantation Immunotherapy, Cellular and Molecular Therapeutics Branch, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD 20892, USA
| | - Angelina Parrizzi
- Laboratory of Transplantation Immunotherapy, Cellular and Molecular Therapeutics Branch, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD 20892, USA
| | - Robert N Reger
- Laboratory of Transplantation Immunotherapy, Cellular and Molecular Therapeutics Branch, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD 20892, USA
| | - Gina M Scurti
- Department of Surgery, Loyola University Chicago, Maywood, IL 60153, USA
| | | | - Richard W Childs
- Laboratory of Transplantation Immunotherapy, Cellular and Molecular Therapeutics Branch, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD 20892, USA
| |
Collapse
|
6
|
Vorri SC, Holl NJ, Leeming M, Apostolova P, Marple A, Ravich JW, Canbaz A, Rahnama R, Choe J, Modi A, Fearnow AD, Walsh ST, Pearce EL, Varadhan R, Bonifant CL. Activation of Cell-Intrinsic Signaling in CAR-T Cells via a Chimeric IL7R Domain. CANCER RESEARCH COMMUNICATIONS 2024; 4:2359-2373. [PMID: 39186002 PMCID: PMC11382189 DOI: 10.1158/2767-9764.crc-24-0286] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/20/2024] [Revised: 08/01/2024] [Accepted: 08/21/2024] [Indexed: 08/27/2024]
Abstract
Chimeric antigen receptor (CAR) T cells can effectively treat leukemias, but sustained antitumor responses can be hindered by a lack of CAR T-cell persistence. Cytotoxic effector T cells are short-lived, and establishment of CAR-T cells with memory to ensure immune surveillance is important. Memory T cells depend on cytokine support, with IL7 activation of the IL7 receptor (IL7R) being critical. However, IL7R surface expression is negatively regulated by exposure to IL7. We aimed to support CAR T-cell persistence by equipping CAR-T cells with a sustained IL7Rα signal. We engineered T cells to constitutively secrete IL7 or to express an anti-acute myeloid leukemia-targeted IL7Rα-chimeric cytokine receptor (CCR) and characterized the phenotype of these cell types. Canonical downstream signaling was activated in CCR-T cells with IL7R activation. When coexpressed with a cytotoxic CAR, functionality of both the CCR and CAR was maintained. We designed hybrid CAR-CCR and noted membrane proximity of the intracellular domains as vital for signaling. These data show cell-intrinsic cytokine support with canonical signaling, and functionality can be provided via expression of an IL7Rα domain whether independently expressed or incorporated into a cytotoxic CAR for use in anticancer therapy. SIGNIFICANCE To improve the phenotype of tumor-directed T-cell therapy, we show that provision of cell-intrinsic IL7R-mediated signaling is preferable to activation of cells with exogenous IL7. We engineer this signaling via independent receptor engineering and incorporation into a CAR and validate maintained antigen-specific cytotoxic activity.
Collapse
MESH Headings
- Humans
- Receptors, Chimeric Antigen/immunology
- Receptors, Chimeric Antigen/genetics
- Receptors, Chimeric Antigen/metabolism
- Signal Transduction
- Immunotherapy, Adoptive/methods
- Interleukin-7/metabolism
- Interleukin-7/genetics
- Receptors, Interleukin-7/metabolism
- Receptors, Interleukin-7/genetics
- Leukemia, Myeloid, Acute/therapy
- Leukemia, Myeloid, Acute/immunology
- Leukemia, Myeloid, Acute/genetics
- Leukemia, Myeloid, Acute/metabolism
- Cell Line, Tumor
- T-Lymphocytes/immunology
- T-Lymphocytes/metabolism
- Interleukin-7 Receptor alpha Subunit
Collapse
Affiliation(s)
- Stamatia C. Vorri
- Department of Oncology, The Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, Maryland.
| | - Natalie J. Holl
- Department of Oncology, The Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, Maryland.
| | - Michael Leeming
- Bio21 Molecular Science and Biotechnology Institute, The University of Melbourne, Victoria, Australia.
| | - Petya Apostolova
- Department of Oncology, The Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, Maryland.
- Bloomberg-Kimmel Institute for Cancer Immunotherapy, Johns Hopkins University School of Medicine, Baltimore, Maryland.
- Division of Hematology, University Hospital Basel, Basel, Switzerland.
| | - Andrew Marple
- Department of Oncology, The Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, Maryland.
| | - Jonas W. Ravich
- Department of Oncology, The Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, Maryland.
| | - Ata Canbaz
- Department of Oncology, The Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, Maryland.
| | - Ruyan Rahnama
- Department of Oncology, The Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, Maryland.
- Department of Pediatrics, Johns Hopkins University School of Medicine, Baltimore, Maryland.
| | - Jun Choe
- Department of Oncology, The Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, Maryland.
- Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, Maryland.
| | - Arjun Modi
- Department of Oncology, The Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, Maryland.
| | - Adam D. Fearnow
- Department of Oncology, The Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, Maryland.
| | - Scott T.R. Walsh
- Chemical Biology Laboratory, National Cancer Institute, National Institutes of Health, Frederick, Maryland.
| | - Erika L. Pearce
- Department of Oncology, The Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, Maryland.
- Bloomberg-Kimmel Institute for Cancer Immunotherapy, Johns Hopkins University School of Medicine, Baltimore, Maryland.
| | - Ravi Varadhan
- Department of Oncology, The Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, Maryland.
| | - Challice L. Bonifant
- Department of Oncology, The Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, Maryland.
- Bloomberg-Kimmel Institute for Cancer Immunotherapy, Johns Hopkins University School of Medicine, Baltimore, Maryland.
- Department of Pediatrics, Johns Hopkins University School of Medicine, Baltimore, Maryland.
| |
Collapse
|
7
|
Chen PH, Raghunandan R, Morrow JS, Katz SG. Finding Your CAR: The Road Ahead for Engineered T Cells. THE AMERICAN JOURNAL OF PATHOLOGY 2024; 194:1409-1423. [PMID: 38697513 PMCID: PMC11284763 DOI: 10.1016/j.ajpath.2024.04.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/18/2023] [Revised: 03/08/2024] [Accepted: 04/02/2024] [Indexed: 05/05/2024]
Abstract
Adoptive cellular therapy using chimeric antigen receptors (CARs) has transformed immunotherapy by engineering T cells to target specific antigens on tumor cells. As the field continues to advance, pathology laboratories will play increasingly essential roles in the complicated multi-step process of CAR T-cell therapy. These include detection of targetable tumor antigens by flow cytometry or immunohistochemistry at the time of disease diagnosis and the isolation and infusion of CAR T cells. Additional roles include: i) detecting antigen loss or heterogeneity that renders resistance to CAR T cells as well as identifying alternative targetable antigens on tumor cells, ii) monitoring the phenotype, persistence, and tumor infiltration properties of CAR T cells and the tumor microenvironment for factors that predict CAR T-cell therapy success, and iii) evaluating side effects and biomarkers of CAR T-cell cytotoxicity such as cytokine release syndrome. This review highlights existing technologies that are applicable to monitoring CAR T-cell persistence, target antigen identification, and loss. Also discussed are emerging technologies that address new challenges such as how to put a brake on CAR T cells. Although pathology laboratories have already provided companion diagnostic tests important in immunotherapy (eg, programmed death-ligand 1, microsatellite instability, and human epidermal growth factor receptor 2 testing), it draws attention to the exciting new translational research opportunities in adoptive cellular therapy.
Collapse
Affiliation(s)
- Po-Han Chen
- Department of Pathology, Yale School of Medicine, New Haven, Connecticut
| | - Rianna Raghunandan
- Department of Pathology, Yale School of Medicine, New Haven, Connecticut
| | - Jon S Morrow
- Department of Pathology, Yale School of Medicine, New Haven, Connecticut
| | - Samuel G Katz
- Department of Pathology, Yale School of Medicine, New Haven, Connecticut.
| |
Collapse
|
8
|
Kandell J, Milian S, Snyder R, Lakshmipathy U. Universal ddPCR-based assay for the determination of lentivirus infectious titer and lenti-modified cell vector copy number. Mol Ther Methods Clin Dev 2023; 31:101120. [PMID: 37841416 PMCID: PMC10568280 DOI: 10.1016/j.omtm.2023.101120] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2023] [Accepted: 09/21/2023] [Indexed: 10/17/2023]
Abstract
The translation of cell-based therapies from research to clinical setting requires robust analytical methods that successfully adhere to current good manufacturing practices and regulatory guidelines. Lentiviral vectors are commonly used for gene delivery to generate genetically modified therapeutic cell products. For some cell therapy products, standardized characterization assays for potency and safety have gained momentum. Translational applications benefit from assays that can be deployed broadly, such as for lentiviral vectors with various transgenes of interest. Development of a universal method to determine lentivirus infectious titer and vector copy number (VCN) of lenti-modified cells was performed using droplet digital PCR (ddPCR). Established methods relied on a ubiquitous lenti-specific target and a housekeeping gene that demonstrated comparability among flow cytometry-based methods. A linearized plasmid control was used to determine assay linearity/range, sensitivity, accuracy, and limits of quantification. Implementing this assay, infectious titer was assessed for various production runs that demonstrated comparability to the flow cytometry titer. The ddPCR assay described here also indicates suitability in the determination of VCN for genetically modified CAR-T cell products. Overall, the development of these universal assays supports the implementation of standardized characterization methods for quality control.
Collapse
Affiliation(s)
- Jennifer Kandell
- Science and Technology, Pharma Services Group, Thermo Fisher Scientific, San Diego, CA 92121, USA
| | - Steven Milian
- Science and Technology, Pharma Services Group, Thermo Fisher Scientific, Alachua, FL 32615, USA
| | - Richard Snyder
- Science and Technology, Pharma Services Group, Thermo Fisher Scientific, Alachua, FL 32615, USA
| | - Uma Lakshmipathy
- Science and Technology, Pharma Services Group, Thermo Fisher Scientific, San Diego, CA 92121, USA
| |
Collapse
|
9
|
Blumenberg V, Busch G, Baumann S, Jitschin R, Iacoboni G, Gallur L, Iraola-Truchuelo J, Hoster E, Winkelmann M, Hellwig K, Schmidt C, Frölich L, Tast B, Hildebrand F, Rejeski K, Dekorsy F, Schmidkonz C, Bäuerle T, Kunz WG, Mougiakakos D, Müller F, von Bergwelt-Baildon M, Barba P, Bücklein VL, Mackensen A, Völkl S, Subklewe M. Early quantification of anti-CD19 CAR T cells by flow cytometry predicts response in R/R DLBCL. Blood Adv 2023; 7:6844-6849. [PMID: 37748131 PMCID: PMC10679803 DOI: 10.1182/bloodadvances.2023010364] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2023] [Revised: 08/02/2023] [Accepted: 09/07/2023] [Indexed: 09/27/2023] Open
Affiliation(s)
- Viktoria Blumenberg
- Department of Medicine III, LMU University Hospital, LMU Munich, Munich, Germany
- Laboratory for Translational Cancer Immunology, LMU Gene Center, LMU Munich, Munich, Germany
- German Cancer Consortium (DKTK), partner site Munich, Munich, Germany
- Bavarian Center for Cancer Research (BZKF), partner site Munich, Munich, Germany
| | - Galina Busch
- Laboratory for Translational Cancer Immunology, LMU Gene Center, LMU Munich, Munich, Germany
| | - Stephan Baumann
- Laboratory for Translational Cancer Immunology, LMU Gene Center, LMU Munich, Munich, Germany
| | - Regina Jitschin
- Department of Internal Medicine 5, Hematology and Oncology, Friedrich-Alexander-Universität Erlangen-Nürnberg and University Hospital Erlangen, Erlangen, Germany
- Department for Hematology and Oncology, Otto-von-Guericke University, Magdeburg, Germany
| | - Gloria Iacoboni
- Department of Hematology, Vall d'Hebron University Hospital, Experimental Hematology, Vall d'Hebron Institute of Oncology, Vall d'Hebron Barcelona Hospital Campus, Barcelona, Spain
- Department of Medicine, Universitat Autònoma of Barcelona, Barcelona, Spain
| | - Laura Gallur
- Department of Hematology, Vall d'Hebron University Hospital, Experimental Hematology, Vall d'Hebron Institute of Oncology, Vall d'Hebron Barcelona Hospital Campus, Barcelona, Spain
- Department of Medicine, Universitat Autònoma of Barcelona, Barcelona, Spain
| | - Josu Iraola-Truchuelo
- Department of Hematology, Vall d'Hebron University Hospital, Experimental Hematology, Vall d'Hebron Institute of Oncology, Vall d'Hebron Barcelona Hospital Campus, Barcelona, Spain
| | - Eva Hoster
- Department of Medicine III, LMU University Hospital, LMU Munich, Munich, Germany
- Institute for Medical Informatics, Biometry and Epidemiology, LMU Munich, Munich, Germany
| | - Michael Winkelmann
- Department of Radiology, LMU University Hospital, LMU Munich, Munich, Germany
| | - Konstantin Hellwig
- Department of Radiology, Friedrich-Alexander-Universität Erlangen-Nürnberg and University Hospital Erlangen, Erlangen, Germany
| | - Christian Schmidt
- Department of Medicine III, LMU University Hospital, LMU Munich, Munich, Germany
| | - Lisa Frölich
- Department of Medicine III, LMU University Hospital, LMU Munich, Munich, Germany
- German Cancer Consortium (DKTK), partner site Munich, Munich, Germany
| | - Benjamin Tast
- Laboratory for Translational Cancer Immunology, LMU Gene Center, LMU Munich, Munich, Germany
| | | | - Kai Rejeski
- Department of Medicine III, LMU University Hospital, LMU Munich, Munich, Germany
- Laboratory for Translational Cancer Immunology, LMU Gene Center, LMU Munich, Munich, Germany
- German Cancer Consortium (DKTK), partner site Munich, Munich, Germany
- Bavarian Center for Cancer Research (BZKF), partner site Munich, Munich, Germany
| | - Franziska Dekorsy
- Department of Nuclear Medicine, LMU University Hospital, LMU Munich, Munich, Germany
| | - Christian Schmidkonz
- Bavarian Center for Cancer Research (BZKF), partner site Munich, Munich, Germany
- Department of Nuclear Medicine, Friedrich-Alexander-Universität Erlangen-Nürnberg and University Hospital Erlangen, Erlangen, Germany
- Institute for Medical Engineering, University of Applied Sciences Amberg-Weiden, Weiden, Germany
| | - Tobias Bäuerle
- Department of Radiology, Friedrich-Alexander-Universität Erlangen-Nürnberg and University Hospital Erlangen, Erlangen, Germany
| | - Wolfgang G. Kunz
- Department of Radiology, LMU University Hospital, LMU Munich, Munich, Germany
| | - Dimitrios Mougiakakos
- Department of Internal Medicine 5, Hematology and Oncology, Friedrich-Alexander-Universität Erlangen-Nürnberg and University Hospital Erlangen, Erlangen, Germany
- Department for Hematology and Oncology, Otto-von-Guericke University, Magdeburg, Germany
- Bavarian Cancer Research Center (BZKF), partner site Erlangen, Erlangen, Germany
| | - Fabian Müller
- Department of Internal Medicine 5, Hematology and Oncology, Friedrich-Alexander-Universität Erlangen-Nürnberg and University Hospital Erlangen, Erlangen, Germany
- Bavarian Cancer Research Center (BZKF), partner site Erlangen, Erlangen, Germany
| | - Michael von Bergwelt-Baildon
- Department of Medicine III, LMU University Hospital, LMU Munich, Munich, Germany
- German Cancer Consortium (DKTK), partner site Munich, Munich, Germany
- Bavarian Center for Cancer Research (BZKF), partner site Munich, Munich, Germany
| | - Pere Barba
- Department of Hematology, Vall d'Hebron University Hospital, Experimental Hematology, Vall d'Hebron Institute of Oncology, Vall d'Hebron Barcelona Hospital Campus, Barcelona, Spain
- Department of Medicine, Universitat Autònoma of Barcelona, Barcelona, Spain
| | - Veit L. Bücklein
- Department of Medicine III, LMU University Hospital, LMU Munich, Munich, Germany
- Laboratory for Translational Cancer Immunology, LMU Gene Center, LMU Munich, Munich, Germany
- Bavarian Center for Cancer Research (BZKF), partner site Munich, Munich, Germany
| | - Andreas Mackensen
- Department of Internal Medicine 5, Hematology and Oncology, Friedrich-Alexander-Universität Erlangen-Nürnberg and University Hospital Erlangen, Erlangen, Germany
- Bavarian Cancer Research Center (BZKF), partner site Erlangen, Erlangen, Germany
| | - Simon Völkl
- Department of Internal Medicine 5, Hematology and Oncology, Friedrich-Alexander-Universität Erlangen-Nürnberg and University Hospital Erlangen, Erlangen, Germany
- Bavarian Cancer Research Center (BZKF), partner site Erlangen, Erlangen, Germany
| | - Marion Subklewe
- Department of Medicine III, LMU University Hospital, LMU Munich, Munich, Germany
- Laboratory for Translational Cancer Immunology, LMU Gene Center, LMU Munich, Munich, Germany
- German Cancer Consortium (DKTK), partner site Munich, Munich, Germany
- Bavarian Center for Cancer Research (BZKF), partner site Munich, Munich, Germany
| |
Collapse
|
10
|
Schubert ML, Schmitt A, Hückelhoven-Krauss A, Neuber B, Kunz A, Waldhoff P, Vonficht D, Yousefian S, Jopp-Saile L, Wang L, Korell F, Keib A, Michels B, Haas D, Sauer T, Derigs P, Kulozik A, Kunz J, Pavel P, Laier S, Wuchter P, Schmier J, Bug G, Lang F, Gökbuget N, Casper J, Görner M, Finke J, Neubauer A, Ringhoffer M, Wolleschak D, Brüggemann M, Haas S, Ho AD, Müller-Tidow C, Dreger P, Schmitt M. Treatment of adult ALL patients with third-generation CD19-directed CAR T cells: results of a pivotal trial. J Hematol Oncol 2023; 16:79. [PMID: 37481608 PMCID: PMC10363324 DOI: 10.1186/s13045-023-01470-0] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2023] [Accepted: 06/20/2023] [Indexed: 07/24/2023] Open
Abstract
BACKGROUND Third-generation chimeric antigen receptor (CAR)-engineered T cells (CARTs) might improve clinical outcome of patients with B cell malignancies. This is the first report on a third-generation CART dose-escalating, phase-1/2 investigator-initiated trial treating adult patients with refractory and/or relapsed (r/r) acute lymphoblastic leukemia (ALL). METHODS Thirteen patients were treated with escalating doses of CD19-directed CARTs between 1 × 106 and 50 × 106 CARTs/m2. Leukapheresis, manufacturing and administration of CARTs were performed in-house. RESULTS For all patients, CART manufacturing was feasible. None of the patients developed any grade of Immune effector cell-associated neurotoxicity syndrome (ICANS) or a higher-grade (≥ grade III) catokine release syndrome (CRS). CART expansion and long-term CART persistence were evident in the peripheral blood (PB) of evaluable patients. At end of study on day 90 after CARTs, ten patients were evaluable for response: Eight patients (80%) achieved a complete remission (CR), including five patients (50%) with minimal residual disease (MRD)-negative CR. Response and outcome were associated with the administered CART dose. At 1-year follow-up, median overall survival was not reached and progression-free survival (PFS) was 38%. Median PFS was reached on day 120. Lack of CD39-expression on memory-like T cells was more frequent in CART products of responders when compared to CART products of non-responders. After CART administration, higher CD8 + and γδ-T cell frequencies, a physiological pattern of immune cells and lower monocyte counts in the PB were associated with response. CONCLUSION In conclusion, third-generation CARTs were associated with promising clinical efficacy and remarkably low procedure-specific toxicity, thereby opening new therapeutic perspectives for patients with r/r ALL. Trial registration This trial was registered at www. CLINICALTRIALS gov as NCT03676504.
Collapse
Affiliation(s)
- Maria-Luisa Schubert
- Department of Internal Medicine V, University Hospital Heidelberg, Im Neuenheimer Feld 410, 69120, Heidelberg, Germany
| | - Anita Schmitt
- Department of Internal Medicine V, University Hospital Heidelberg, Im Neuenheimer Feld 410, 69120, Heidelberg, Germany
| | - Angela Hückelhoven-Krauss
- Department of Internal Medicine V, University Hospital Heidelberg, Im Neuenheimer Feld 410, 69120, Heidelberg, Germany
| | - Brigitte Neuber
- Department of Internal Medicine V, University Hospital Heidelberg, Im Neuenheimer Feld 410, 69120, Heidelberg, Germany
| | - Alexander Kunz
- Department of Internal Medicine V, University Hospital Heidelberg, Im Neuenheimer Feld 410, 69120, Heidelberg, Germany
| | - Philip Waldhoff
- Department of Internal Medicine V, University Hospital Heidelberg, Im Neuenheimer Feld 410, 69120, Heidelberg, Germany
| | - Dominik Vonficht
- Heidelberg Institute for Stem Cell Technology and Experimental Medicine (HI-STEM gGmbH), Heidelberg, Germany
- Division of Stem Cells and Cancer, Deutsches Krebsforschungszentrum (DKFZ) and DKFZ-ZMBH Alliance, Heidelberg, Germany
- Faculty of Biosciences, Heidelberg University, Heidelberg, Germany
| | - Schayan Yousefian
- Berlin Institute of Health (BIH) at Charité - Universitätsmedizin Berlin, Berlin, Germany
- Berlin Institute for Medical Systems Biology, Max Delbrück Center for Molecular Medicine in the Helmholtz Association, Berlin, Germany
- Department of Hematology, Oncology and Tumor Immunology, Charité University Medicine, Berlin, Germany
| | - Lea Jopp-Saile
- Heidelberg Institute for Stem Cell Technology and Experimental Medicine (HI-STEM gGmbH), Heidelberg, Germany
- Division of Stem Cells and Cancer, Deutsches Krebsforschungszentrum (DKFZ) and DKFZ-ZMBH Alliance, Heidelberg, Germany
- Faculty of Biosciences, Heidelberg University, Heidelberg, Germany
| | - Lei Wang
- Department of Internal Medicine V, University Hospital Heidelberg, Im Neuenheimer Feld 410, 69120, Heidelberg, Germany
| | - Felix Korell
- Department of Internal Medicine V, University Hospital Heidelberg, Im Neuenheimer Feld 410, 69120, Heidelberg, Germany
| | - Anna Keib
- Department of Internal Medicine V, University Hospital Heidelberg, Im Neuenheimer Feld 410, 69120, Heidelberg, Germany
| | - Birgit Michels
- Department of Internal Medicine V, University Hospital Heidelberg, Im Neuenheimer Feld 410, 69120, Heidelberg, Germany
| | - Dominik Haas
- Department of Internal Medicine V, University Hospital Heidelberg, Im Neuenheimer Feld 410, 69120, Heidelberg, Germany
| | - Tim Sauer
- Department of Internal Medicine V, University Hospital Heidelberg, Im Neuenheimer Feld 410, 69120, Heidelberg, Germany
| | - Patrick Derigs
- Department of Internal Medicine V, University Hospital Heidelberg, Im Neuenheimer Feld 410, 69120, Heidelberg, Germany
| | - Andreas Kulozik
- Department of Pediatric Hematology, Oncology and Immunology, University Hospital Heidelberg, Heidelberg, Germany
| | - Joachim Kunz
- Department of Pediatric Hematology, Oncology and Immunology, University Hospital Heidelberg, Heidelberg, Germany
| | - Petra Pavel
- Institute for Clinical Transfusion Medicine and Cell Therapy (IKTZ), German Red Cross Blood Service Baden-Württemberg-Hessen, Heidelberg, Germany
| | - Sascha Laier
- Institute for Clinical Transfusion Medicine and Cell Therapy (IKTZ), German Red Cross Blood Service Baden-Württemberg-Hessen, Heidelberg, Germany
| | - Patrick Wuchter
- Institute of Transfusion Medicine and Immunology, Medical Faculty Mannheim, of the Heidelberg University, German Red Cross Blood Service Baden-Württemberg - Hessen, Mannheim, Germany
| | | | - Gesine Bug
- Department of Internal Medicine II, University Hospital Frankfurt, Frankfurt, Germany
| | - Fabian Lang
- Department of Internal Medicine II, University Hospital Frankfurt, Frankfurt, Germany
| | - Nicola Gökbuget
- Department of Internal Medicine II, University Hospital Frankfurt, Frankfurt, Germany
| | - Jochen Casper
- Department of Hematology and Oncology, University Hospital Oldenburg, Oldenburg, Germany
| | - Martin Görner
- Department of Hematology and Oncology, Hospital Bielefeld, Bielefeld, Germany
| | - Jürgen Finke
- Department of Internal Medicine I, University Hospital Freiburg, Freiburg, Germany
| | - Andreas Neubauer
- Department of Hematology, Oncology and Immunology, University Hospital Giessen und Marburg, Marburg, Germany
| | | | - Denise Wolleschak
- Department of Hematology and Oncology, Center of Internal Medicine, Otto-von-Guericke University Medical Center, Magdeburg, Germany
| | - Monika Brüggemann
- Department of Internal Medicine II, University Hospital Kiel, Kiel, Germany
| | - Simon Haas
- Heidelberg Institute for Stem Cell Technology and Experimental Medicine (HI-STEM gGmbH), Heidelberg, Germany
- Division of Stem Cells and Cancer, Deutsches Krebsforschungszentrum (DKFZ) and DKFZ-ZMBH Alliance, Heidelberg, Germany
- Berlin Institute of Health (BIH) at Charité - Universitätsmedizin Berlin, Berlin, Germany
- Berlin Institute for Medical Systems Biology, Max Delbrück Center for Molecular Medicine in the Helmholtz Association, Berlin, Germany
- Department of Hematology, Oncology and Tumor Immunology, Charité University Medicine, Berlin, Germany
- German Cancer Consortium (DKTK) and German Cancer Research Center (DKFZ)/National Center for Tumor Diseases (NCT), Heidelberg, Germany
| | - Anthony D Ho
- Department of Internal Medicine V, University Hospital Heidelberg, Im Neuenheimer Feld 410, 69120, Heidelberg, Germany
- German Cancer Consortium (DKTK) and German Cancer Research Center (DKFZ)/National Center for Tumor Diseases (NCT), Heidelberg, Germany
| | - Carsten Müller-Tidow
- Department of Internal Medicine V, University Hospital Heidelberg, Im Neuenheimer Feld 410, 69120, Heidelberg, Germany
- German Cancer Consortium (DKTK) and German Cancer Research Center (DKFZ)/National Center for Tumor Diseases (NCT), Heidelberg, Germany
| | - Peter Dreger
- Department of Internal Medicine V, University Hospital Heidelberg, Im Neuenheimer Feld 410, 69120, Heidelberg, Germany
- German Cancer Consortium (DKTK) and German Cancer Research Center (DKFZ)/National Center for Tumor Diseases (NCT), Heidelberg, Germany
| | - Michael Schmitt
- Department of Internal Medicine V, University Hospital Heidelberg, Im Neuenheimer Feld 410, 69120, Heidelberg, Germany.
- German Cancer Consortium (DKTK) and German Cancer Research Center (DKFZ)/National Center for Tumor Diseases (NCT), Heidelberg, Germany.
| |
Collapse
|
11
|
Murphy LA, Marians RC, Miller K, Brenton MD, Mallo RLV, Kohler ME, Fry TJ, Winters AC. Digital polymerase chain reaction strategies for accurate and precise detection of vector copy number in chimeric antigen receptor T-cell products. Cytotherapy 2023; 25:94-102. [PMID: 36253252 PMCID: PMC10123956 DOI: 10.1016/j.jcyt.2022.09.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2022] [Revised: 08/31/2022] [Accepted: 09/14/2022] [Indexed: 12/27/2022]
Abstract
BACKGROUND AIMS Vector copy number (VCN), an average quantification of transgene copies unique to a chimeric antigen receptor (CAR) T-cell product, is a characteristic that must be reported prior to patient administration, as high VCN increases the risk of insertional mutagenesis. Historically, VCN assessment in CAR T-cell products has been performed via quantitative polymerase chain reaction (qPCR). qPCR is reliable along a broad range of concentrations, but quantification requires use of a standard curve and precision is limited. Digital PCR (dPCR) methods were developed for absolute quantification of target sequences by counting nucleic acid molecules encapsulated in discrete, volumetrically defined partitions. Advantages of dPCR compared with qPCR include simplicity, reproducibility, sensitivity and lack of dependency on a standard curve for definitive quantification. In the present study, the authors describe a dPCR assay developed for analysis of the novel bicistronic CD19 × CD22 CAR T-cell construct. METHODS The authors compared the performance of the dPCR assay with qPCR on both the QX200 droplet dPCR (ddPCR) system (Bio-Rad Laboratories, Inc, Hercules, CA, USA) and the QIAcuity nanoplate-based dPCR (ndPCR) system (QIAGEN Sciences, Inc, Germantown, MD, USA). The primer-probe assay was validated with qPCR, ndPCR and ddPCR using patient samples from pre-clinical CAR T-cell manufacturing production runs as well as Jurkat cell subclones, which stably express this bicistronic CAR construct. RESULTS ddPCR confirmed the specificity of this assay to detect only the bicistronic CAR product. Additionally, the authors' assay gave accurate, precise and reproducible CAR T-cell VCN measurements across qPCR, ndPCR and ddPCR modalities. CONCLUSIONS The authors demonstrate that dPCR strategies can be utilized for absolute quantification of CAR transgenes and VCN measurements, with improved test-retest reliability, and that specific assays can be developed for detection of unique constructs.
Collapse
Affiliation(s)
- Lindsey A Murphy
- Center for Cancer and Blood Disorders, Children's Hospital Colorado and Department of Pediatrics, University of Colorado Anschutz Medical Campus, Aurora, Colorado, USA
| | - Russell C Marians
- Charles C. Gates Biomanufacturing Facility, University of Colorado Anschutz Medical Campus, Aurora, Colorado, USA
| | - Kristen Miller
- Center for Cancer and Blood Disorders, Children's Hospital Colorado and Department of Pediatrics, University of Colorado Anschutz Medical Campus, Aurora, Colorado, USA
| | - Matthew D Brenton
- Charles C. Gates Biomanufacturing Facility, University of Colorado Anschutz Medical Campus, Aurora, Colorado, USA
| | - Rebecca L V Mallo
- Charles C. Gates Biomanufacturing Facility, University of Colorado Anschutz Medical Campus, Aurora, Colorado, USA
| | - M Eric Kohler
- Center for Cancer and Blood Disorders, Children's Hospital Colorado and Department of Pediatrics, University of Colorado Anschutz Medical Campus, Aurora, Colorado, USA
| | - Terry J Fry
- Center for Cancer and Blood Disorders, Children's Hospital Colorado and Department of Pediatrics, University of Colorado Anschutz Medical Campus, Aurora, Colorado, USA
| | - Amanda C Winters
- Center for Cancer and Blood Disorders, Children's Hospital Colorado and Department of Pediatrics, University of Colorado Anschutz Medical Campus, Aurora, Colorado, USA.
| |
Collapse
|
12
|
García-Calderón CB, Sierro-Martínez B, García-Guerrero E, Sanoja-Flores L, Muñoz-García R, Ruiz-Maldonado V, Jimenez-Leon MR, Delgado-Serrano J, Molinos-Quintana Á, Guijarro-Albaladejo B, Carrasco-Brocal I, Lucena JM, García-Lozano JR, Blázquez-Goñi C, Reguera-Ortega JL, González-Escribano MF, Reinoso-Segura M, Briones J, Pérez-Simón JA, Caballero-Velázquez T. Monitoring of kinetics and exhaustion markers of circulating CAR-T cells as early predictive factors in patients with B-cell malignancies. Front Immunol 2023; 14:1152498. [PMID: 37122702 PMCID: PMC10140355 DOI: 10.3389/fimmu.2023.1152498] [Citation(s) in RCA: 21] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2023] [Accepted: 03/31/2023] [Indexed: 05/02/2023] Open
Abstract
Purpose CAR-T cell therapy has proven to be a disruptive treatment in the hematology field, however, less than 50% of patients maintain long-term response and early predictors of outcome are still inconsistently defined. Here, we aimed to optimize the detection of CD19 CAR-T cells in blood and to identify phenotypic features as early biomarkers associated with toxicity and outcomes. Experimental design In this study, monitoring by flow cytometry and digital PCR (dPCR), and immunophenotypic characterization of circulating CAR-T cells from 48 patients treated with Tisa-cel or Axi-cel was performed. Results Validation of the flow cytometry reagent for the detection of CAR-T cells in blood revealed CD19 protein conjugated with streptavidin as the optimal detection method. Kinetics of CAR-T cell expansion in blood confirmed median day of peak expansion at seven days post-infusion by both flow cytometry and digital PCR. Circulating CAR-T cells showed an activated, proliferative, and exhausted phenotype at the time of peak expansion. Patients with increased expansion showed more severe CRS and ICANs. Immunophenotypic characterization of CAR-T cells at the peak expansion identified the increased expression of co-inhibitory molecules PD1 and LAG3 and reduced levels of the cytotoxicity marker CD107a as predictors of a better long-term disease control. Conclusions These data show the importance of CAR-T cells in vivo monitoring and identify the expression of PD1LAG3 and CD107a as early biomarkers of long-term disease control after CAR-T cell therapy.
Collapse
Affiliation(s)
- Clara Beatriz García-Calderón
- Servicio de Hematología, Hospital Universitario Virgen del Rocío, Instituto de Biomedicina de Sevilla, (IBIS/CSIC), Universidad de Sevilla, Sevilla, Spain
| | - Belén Sierro-Martínez
- Servicio de Hematología, Hospital Universitario Virgen del Rocío, Instituto de Biomedicina de Sevilla, (IBIS/CSIC), Universidad de Sevilla, Sevilla, Spain
| | - Estefanía García-Guerrero
- Servicio de Hematología, Hospital Universitario Virgen del Rocío, Instituto de Biomedicina de Sevilla, (IBIS/CSIC), Universidad de Sevilla, Sevilla, Spain
- *Correspondence: Estefanía García-Guerrero,
| | - Luzalba Sanoja-Flores
- Servicio de Hematología, Hospital Universitario Virgen del Rocío, Instituto de Biomedicina de Sevilla, (IBIS/CSIC), Universidad de Sevilla, Sevilla, Spain
| | - Raquel Muñoz-García
- Servicio de Inmunología, Instituto de Biomedicina de Sevilla (IBiS), Hospital Universitario Virgen del Rocío, Centro Superior de Investigaciones Científicas (CSIC), Universidad de Sevilla, Sevilla, Spain
| | - Victoria Ruiz-Maldonado
- Servicio de Hematología, Hospital Universitario Virgen del Rocío, Instituto de Biomedicina de Sevilla, (IBIS/CSIC), Universidad de Sevilla, Sevilla, Spain
| | - María Reyes Jimenez-Leon
- Servicio de Hematología, Hospital Universitario Virgen del Rocío, Instituto de Biomedicina de Sevilla, (IBIS/CSIC), Universidad de Sevilla, Sevilla, Spain
| | - Javier Delgado-Serrano
- Servicio de Hematología, Hospital Universitario Virgen del Rocío, Instituto de Biomedicina de Sevilla, (IBIS/CSIC), Universidad de Sevilla, Sevilla, Spain
| | - Águeda Molinos-Quintana
- Servicio de Hematología, Hospital Universitario Virgen del Rocío, Instituto de Biomedicina de Sevilla, (IBIS/CSIC), Universidad de Sevilla, Sevilla, Spain
| | - Beatriz Guijarro-Albaladejo
- Servicio de Hematología, Hospital Universitario Virgen del Rocío, Instituto de Biomedicina de Sevilla, (IBIS/CSIC), Universidad de Sevilla, Sevilla, Spain
| | - Inmaculada Carrasco-Brocal
- Servicio de Hematología, Hospital Universitario Virgen del Rocío, Instituto de Biomedicina de Sevilla, (IBIS/CSIC), Universidad de Sevilla, Sevilla, Spain
| | - José-Manuel Lucena
- Servicio de Inmunología, Instituto de Biomedicina de Sevilla (IBiS), Hospital Universitario Virgen del Rocío, Centro Superior de Investigaciones Científicas (CSIC), Universidad de Sevilla, Sevilla, Spain
| | - José-Raúl García-Lozano
- Servicio de Inmunología, Instituto de Biomedicina de Sevilla (IBiS), Hospital Universitario Virgen del Rocío, Centro Superior de Investigaciones Científicas (CSIC), Universidad de Sevilla, Sevilla, Spain
| | - Cristina Blázquez-Goñi
- Servicio de Hematología, Hospital Universitario Virgen del Rocío, Instituto de Biomedicina de Sevilla, (IBIS/CSIC), Universidad de Sevilla, Sevilla, Spain
| | - Juan Luis Reguera-Ortega
- Servicio de Hematología, Hospital Universitario Virgen del Rocío, Instituto de Biomedicina de Sevilla, (IBIS/CSIC), Universidad de Sevilla, Sevilla, Spain
| | - María-Francisca González-Escribano
- Servicio de Inmunología, Instituto de Biomedicina de Sevilla (IBiS), Hospital Universitario Virgen del Rocío, Centro Superior de Investigaciones Científicas (CSIC), Universidad de Sevilla, Sevilla, Spain
| | - Marta Reinoso-Segura
- Servicio de Hematología, Hospital Universitario Virgen del Rocío, Instituto de Biomedicina de Sevilla, (IBIS/CSIC), Universidad de Sevilla, Sevilla, Spain
| | - Javier Briones
- Hematology Service, Hospital de la Santa Creu i Sant Pau, Barcelona, Spain
| | - José Antonio Pérez-Simón
- Servicio de Hematología, Hospital Universitario Virgen del Rocío, Instituto de Biomedicina de Sevilla, (IBIS/CSIC), Universidad de Sevilla, Sevilla, Spain
| | - Teresa Caballero-Velázquez
- Servicio de Hematología, Hospital Universitario Virgen del Rocío, Instituto de Biomedicina de Sevilla, (IBIS/CSIC), Universidad de Sevilla, Sevilla, Spain
| |
Collapse
|
13
|
Mueller KP, Piscopo NJ, Forsberg MH, Saraspe LA, Das A, Russell B, Smerchansky M, Cappabianca D, Shi L, Shankar K, Sarko L, Khajanchi N, La Vonne Denne N, Ramamurthy A, Ali A, Lazzarotto CR, Tsai SQ, Capitini CM, Saha K. Production and characterization of virus-free, CRISPR-CAR T cells capable of inducing solid tumor regression. J Immunother Cancer 2022; 10:e004446. [PMID: 36382633 PMCID: PMC9454086 DOI: 10.1136/jitc-2021-004446] [Citation(s) in RCA: 26] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 08/04/2022] [Indexed: 12/14/2022] Open
Abstract
BACKGROUND Chimeric antigen receptor (CAR) T cells have demonstrated high clinical response rates against hematological malignancies (e.g., CD19+ cancers) but have shown limited activity in patients with solid tumors. Recent work showed that precise insertion of a CAR at a defined locus improves treatment outcomes in the context of a CD19 CAR; however, it is unclear if such a strategy could also affect outcomes in solid tumors. Furthermore, CAR manufacturing generally relies on viral vectors for gene delivery, which comprise a complex and resource-intensive part of the manufacturing supply chain. METHODS Anti-GD2 CAR T cells were generated using CRISPR/Cas9 within 9 days using recombinant Cas9 protein and nucleic acids, without any viral vectors. The CAR was specifically targeted to the T cell receptor alpha constant gene (TRAC). T cell products were characterized at the level of the genome, transcriptome, proteome, and secretome using CHANGE-seq, targeted next-generation sequencing, scRNA-seq, spectral cytometry, and ELISA assays, respectively. Functionality was evaluated in vivo in an NSG™ xenograft neuroblastoma model. RESULTS In comparison to retroviral CAR T cells, virus-free CRISPR CAR (VFC-CAR) T cells exhibit TRAC-targeted genomic integration of the CAR transgene, elevation of transcriptional and protein characteristics associated with a memory-like phenotype, and low tonic signaling prior to infusion arising in part from the knockout of the T cell receptor. On exposure to the GD2 target antigen, anti-GD2 VFC-CAR T cells exhibit specific cytotoxicity against GD2+ cells in vitro and induce solid tumor regression in vivo. VFC-CAR T cells demonstrate robust homing and persistence and decreased exhaustion relative to retroviral CAR T cells against a human neuroblastoma xenograft model. CONCLUSIONS This study leverages virus-free genome editing technology to generate CAR T cells featuring a TRAC-targeted CAR, which could inform manufacturing of CAR T cells to treat cancers, including solid tumors.
Collapse
Affiliation(s)
- Katherine P Mueller
- Department of Biomedical Engineering, University of Wisconsin-Madison, Madison, Wisconsin, USA
- Wisconsin Institute for Discovery, University of Wisconsin-Madison, Madison, Wisconsin, USA
| | - Nicole J Piscopo
- Department of Biomedical Engineering, University of Wisconsin-Madison, Madison, Wisconsin, USA
- Wisconsin Institute for Discovery, University of Wisconsin-Madison, Madison, Wisconsin, USA
| | - Matthew H Forsberg
- Department of Pediatrics, University of Wisconsin School of Medicine and Public Health, Madison, Wisconsin, USA
| | - Louise A Saraspe
- Wisconsin Institute for Discovery, University of Wisconsin-Madison, Madison, Wisconsin, USA
| | - Amritava Das
- Wisconsin Institute for Discovery, University of Wisconsin-Madison, Madison, Wisconsin, USA
| | - Brittany Russell
- Wisconsin Institute for Discovery, University of Wisconsin-Madison, Madison, Wisconsin, USA
| | - Madeline Smerchansky
- Department of Biomedical Engineering, University of Wisconsin-Madison, Madison, Wisconsin, USA
- Wisconsin Institute for Discovery, University of Wisconsin-Madison, Madison, Wisconsin, USA
| | - Dan Cappabianca
- Department of Biomedical Engineering, University of Wisconsin-Madison, Madison, Wisconsin, USA
- Wisconsin Institute for Discovery, University of Wisconsin-Madison, Madison, Wisconsin, USA
| | - Lei Shi
- Department of Pediatrics, University of Wisconsin School of Medicine and Public Health, Madison, Wisconsin, USA
| | - Keerthana Shankar
- Department of Biomedical Engineering, University of Wisconsin-Madison, Madison, Wisconsin, USA
- Wisconsin Institute for Discovery, University of Wisconsin-Madison, Madison, Wisconsin, USA
| | - Lauren Sarko
- Department of Biomedical Engineering, University of Wisconsin-Madison, Madison, Wisconsin, USA
- Wisconsin Institute for Discovery, University of Wisconsin-Madison, Madison, Wisconsin, USA
| | - Namita Khajanchi
- Department of Biomedical Engineering, University of Wisconsin-Madison, Madison, Wisconsin, USA
- Wisconsin Institute for Discovery, University of Wisconsin-Madison, Madison, Wisconsin, USA
| | - Nina La Vonne Denne
- Department of Biomedical Engineering, University of Wisconsin-Madison, Madison, Wisconsin, USA
- Wisconsin Institute for Discovery, University of Wisconsin-Madison, Madison, Wisconsin, USA
| | - Apoorva Ramamurthy
- Department of Biomedical Engineering, University of Wisconsin-Madison, Madison, Wisconsin, USA
- Wisconsin Institute for Discovery, University of Wisconsin-Madison, Madison, Wisconsin, USA
| | - Adeela Ali
- Department of Pediatrics, University of Wisconsin School of Medicine and Public Health, Madison, Wisconsin, USA
| | - Cicera R Lazzarotto
- Department of Hematology, St Jude Children's Research Hospital, Memphis, Tennessee, USA
| | - Shengdar Q Tsai
- Department of Hematology, St Jude Children's Research Hospital, Memphis, Tennessee, USA
| | - Christian M Capitini
- Department of Pediatrics, University of Wisconsin School of Medicine and Public Health, Madison, Wisconsin, USA
| | - Krishanu Saha
- Department of Biomedical Engineering, University of Wisconsin-Madison, Madison, Wisconsin, USA
- Wisconsin Institute for Discovery, University of Wisconsin-Madison, Madison, Wisconsin, USA
| |
Collapse
|
14
|
Clinically Applicable Assessment of Tisagenlecleucel CAR T Cell Treatment by Digital Droplet PCR for Copy Number Variant Assessment. Int J Mol Sci 2022; 23:ijms23147573. [PMID: 35886920 PMCID: PMC9322953 DOI: 10.3390/ijms23147573] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2022] [Revised: 07/01/2022] [Accepted: 07/04/2022] [Indexed: 12/04/2022] Open
Abstract
Chimeric antigen receptor (CAR) T cell therapy is an innovative immunotherapy for treating cancers in both children and adults with proven utility in numerous clinical trials. Significantly, some CAR T cell therapies have now been approved by relevant national regulatory bodies across numerous countries for clinical therapeutic use outside of clinical trials. One such recently licensed product is tisagenlecleucel, a CAR T therapy approved for the treatment of B-cell acute lymphoblastic leukemia (B-ALL) using autologous T cells from the patient. The genetically engineered T cells target a protein called CD19, common to B cells, through a CAR incorporating a 4-1BB costimulatory domain to improve response. Since tisagenlecleucel is now a standard of care treatment for B-ALL, it is clinically essential to be able to accurately monitor these CAR T cells in patients. Assessment of the copy number variant (CNV) of the CAR T cell products allows this within a clinically acceptable timeframe for optimal patient benefit. However, no standardized method with high reproducibility and efficiency has been described within a routine clinical laboratory setting. Here, we demonstrated a novel digital droplet PCR (ddPCR)-based methodology for the study of CNV (ddPCR-CNV) in 4-1BB CD19-specific CAR T cells with universal applicability across clinical diagnostic laboratories.
Collapse
|
15
|
Schubert ML, Berger C, Kunz A, Schmitt A, Badbaran A, Neuber B, Zeschke S, Wang L, Riecken K, Hückelhoven‑Krauss A, Müller I, Müller‑Tidow C, Dreger P, Kröger N, Ayuk F, Schmitt M, Fehse B. Comparison of single copy gene‑based duplex quantitative PCR and digital droplet PCR for monitoring of expansion of CD19‑directed CAR T cells in treated patients. Int J Oncol 2022; 60:48. [PMID: 35294040 PMCID: PMC8973917 DOI: 10.3892/ijo.2022.5338] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2021] [Accepted: 02/11/2022] [Indexed: 11/06/2022] Open
Affiliation(s)
- Maria-Luisa Schubert
- Department of Internal Medicine V (Hematology/Oncology/Rheumatology), University Hospital Heidelberg, D‑69120 Heidelberg, Germany
| | - Carolina Berger
- Department of Stem Cell Transplantation, University Medical Centre Hamburg‑Eppendorf, D‑20246 Hamburg, Germany
| | - Alexander Kunz
- Department of Internal Medicine V (Hematology/Oncology/Rheumatology), University Hospital Heidelberg, D‑69120 Heidelberg, Germany
| | - Anita Schmitt
- Department of Internal Medicine V (Hematology/Oncology/Rheumatology), University Hospital Heidelberg, D‑69120 Heidelberg, Germany
| | - Anita Badbaran
- Department of Stem Cell Transplantation, University Medical Centre Hamburg‑Eppendorf, D‑20246 Hamburg, Germany
| | - Brigitte Neuber
- Department of Internal Medicine V (Hematology/Oncology/Rheumatology), University Hospital Heidelberg, D‑69120 Heidelberg, Germany
| | - Silke Zeschke
- Department of Stem Cell Transplantation, University Medical Centre Hamburg‑Eppendorf, D‑20246 Hamburg, Germany
| | - Lei Wang
- Department of Internal Medicine V (Hematology/Oncology/Rheumatology), University Hospital Heidelberg, D‑69120 Heidelberg, Germany
| | - Kristoffer Riecken
- Department of Stem Cell Transplantation, University Medical Centre Hamburg‑Eppendorf, D‑20246 Hamburg, Germany
| | - Angela Hückelhoven‑Krauss
- Department of Internal Medicine V (Hematology/Oncology/Rheumatology), University Hospital Heidelberg, D‑69120 Heidelberg, Germany
| | - Ingo Müller
- Department of Pediatric Hematology and Oncology, University Medical Centre Hamburg‑Eppendorf, D‑20246 Hamburg, Germany
| | - Carsten Müller‑Tidow
- Department of Internal Medicine V (Hematology/Oncology/Rheumatology), University Hospital Heidelberg, D‑69120 Heidelberg, Germany
| | - Peter Dreger
- Department of Internal Medicine V (Hematology/Oncology/Rheumatology), University Hospital Heidelberg, D‑69120 Heidelberg, Germany
| | - Nicolaus Kröger
- Department of Stem Cell Transplantation, University Medical Centre Hamburg‑Eppendorf, D‑20246 Hamburg, Germany
| | - Francis Ayuk
- Department of Stem Cell Transplantation, University Medical Centre Hamburg‑Eppendorf, D‑20246 Hamburg, Germany
| | - Michael Schmitt
- Department of Internal Medicine V (Hematology/Oncology/Rheumatology), University Hospital Heidelberg, D‑69120 Heidelberg, Germany
| | - Boris Fehse
- Department of Stem Cell Transplantation, University Medical Centre Hamburg‑Eppendorf, D‑20246 Hamburg, Germany
| |
Collapse
|
16
|
Reichman A, Kunz A, Joedicke JJ, Höpken UE, Keib A, Neuber B, Sedloev D, Wang L, Jiang G, Hückelhoven-Krauss A, Eberhardt F, Müller-Tidow C, Wermke M, Rehm A, Schmitt M, Schmitt A. Comparison of FACS and PCR for Detection of BCMA-CAR-T Cells. Int J Mol Sci 2022; 23:ijms23020903. [PMID: 35055086 PMCID: PMC8777942 DOI: 10.3390/ijms23020903] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2021] [Revised: 01/11/2022] [Accepted: 01/12/2022] [Indexed: 02/01/2023] Open
Abstract
Chimeric-antigen-receptor (CAR)-T-cell therapy is already widely used to treat patients who are relapsed or refractory to chemotherapy, antibodies, or stem-cell transplantation. Multiple myeloma still constitutes an incurable disease. CAR-T-cell therapy that targets BCMA (B-cell maturation antigen) is currently revolutionizing the treatment of those patients. To monitor and improve treatment outcomes, methods to detect CAR-T cells in human peripheral blood are highly desirable. In this study, three different detection reagents for staining BCMA-CAR-T cells by flow cytometry were compared. Moreover, a quantitative polymerase chain reaction (qPCR) to detect BCMA-CAR-T cells was established. By applying a cell-titration experiment of BCMA-CAR-T cells, both methods were compared head-to-head. In flow-cytometric analysis, the detection reagents used in this study could all detect BCMA-CAR-T cells at a similar level. The results of false-positive background staining differed as follows (standard deviation): the BCMA-detection reagent used on the control revealed a background staining of 0.04% (±0.02%), for the PE-labeled human BCMA peptide it was 0.25% (±0.06%) and for the polyclonal anti-human IgG antibody it was 7.2% (±9.2%). The ability to detect BCMA-CAR-T cells down to a concentration of 0.4% was similar for qPCR and flow cytometry. The qPCR could detect even lower concentrations (0.02–0.01%). In summary, BCMA-CAR-T-cell monitoring can be reliably performed by both flow cytometry and qPCR. In flow cytometry, reagents with low background staining should be preferred.
Collapse
Affiliation(s)
- Avinoam Reichman
- Department of Hematology, Oncology and Rheumatology, Heidelberg University Hospital, Im Neuenheimer Feld 410, 69120 Heidelberg, Germany; (A.R.); (A.K.); (A.K.); (B.N.); (D.S.); (L.W.); (G.J.); (A.H.-K.); (F.E.); (C.M.-T.); (M.S.)
| | - Alexander Kunz
- Department of Hematology, Oncology and Rheumatology, Heidelberg University Hospital, Im Neuenheimer Feld 410, 69120 Heidelberg, Germany; (A.R.); (A.K.); (A.K.); (B.N.); (D.S.); (L.W.); (G.J.); (A.H.-K.); (F.E.); (C.M.-T.); (M.S.)
| | - Jara J. Joedicke
- Department of Microenvironmental Regulation in Autoimmunity and Cancer, Max-Delbrück Center for Molecular Medicine (MDC), Robert-Rössle-Str. 10, 13125 Berlin-Buch, Germany; (J.J.J.); (A.R.)
| | - Uta E. Höpken
- Department of Translational Tumor Immunology, Max-Delbrück Center for Molecular Medicine (MDC), Robert-Rössle-Str. 10, 13125 Berlin-Buch, Germany;
| | - Anna Keib
- Department of Hematology, Oncology and Rheumatology, Heidelberg University Hospital, Im Neuenheimer Feld 410, 69120 Heidelberg, Germany; (A.R.); (A.K.); (A.K.); (B.N.); (D.S.); (L.W.); (G.J.); (A.H.-K.); (F.E.); (C.M.-T.); (M.S.)
| | - Brigitte Neuber
- Department of Hematology, Oncology and Rheumatology, Heidelberg University Hospital, Im Neuenheimer Feld 410, 69120 Heidelberg, Germany; (A.R.); (A.K.); (A.K.); (B.N.); (D.S.); (L.W.); (G.J.); (A.H.-K.); (F.E.); (C.M.-T.); (M.S.)
| | - David Sedloev
- Department of Hematology, Oncology and Rheumatology, Heidelberg University Hospital, Im Neuenheimer Feld 410, 69120 Heidelberg, Germany; (A.R.); (A.K.); (A.K.); (B.N.); (D.S.); (L.W.); (G.J.); (A.H.-K.); (F.E.); (C.M.-T.); (M.S.)
| | - Lei Wang
- Department of Hematology, Oncology and Rheumatology, Heidelberg University Hospital, Im Neuenheimer Feld 410, 69120 Heidelberg, Germany; (A.R.); (A.K.); (A.K.); (B.N.); (D.S.); (L.W.); (G.J.); (A.H.-K.); (F.E.); (C.M.-T.); (M.S.)
| | - Genqiao Jiang
- Department of Hematology, Oncology and Rheumatology, Heidelberg University Hospital, Im Neuenheimer Feld 410, 69120 Heidelberg, Germany; (A.R.); (A.K.); (A.K.); (B.N.); (D.S.); (L.W.); (G.J.); (A.H.-K.); (F.E.); (C.M.-T.); (M.S.)
| | - Angela Hückelhoven-Krauss
- Department of Hematology, Oncology and Rheumatology, Heidelberg University Hospital, Im Neuenheimer Feld 410, 69120 Heidelberg, Germany; (A.R.); (A.K.); (A.K.); (B.N.); (D.S.); (L.W.); (G.J.); (A.H.-K.); (F.E.); (C.M.-T.); (M.S.)
| | - Franziska Eberhardt
- Department of Hematology, Oncology and Rheumatology, Heidelberg University Hospital, Im Neuenheimer Feld 410, 69120 Heidelberg, Germany; (A.R.); (A.K.); (A.K.); (B.N.); (D.S.); (L.W.); (G.J.); (A.H.-K.); (F.E.); (C.M.-T.); (M.S.)
| | - Carsten Müller-Tidow
- Department of Hematology, Oncology and Rheumatology, Heidelberg University Hospital, Im Neuenheimer Feld 410, 69120 Heidelberg, Germany; (A.R.); (A.K.); (A.K.); (B.N.); (D.S.); (L.W.); (G.J.); (A.H.-K.); (F.E.); (C.M.-T.); (M.S.)
| | - Martin Wermke
- NCT/UCC Early Clinical Trial Unit (ECTU), Medical Faculty C.-G. Carus, Technical University Dresden, Fetscherstraße 74, 01307 Dresden, Germany;
| | - Armin Rehm
- Department of Microenvironmental Regulation in Autoimmunity and Cancer, Max-Delbrück Center for Molecular Medicine (MDC), Robert-Rössle-Str. 10, 13125 Berlin-Buch, Germany; (J.J.J.); (A.R.)
| | - Michael Schmitt
- Department of Hematology, Oncology and Rheumatology, Heidelberg University Hospital, Im Neuenheimer Feld 410, 69120 Heidelberg, Germany; (A.R.); (A.K.); (A.K.); (B.N.); (D.S.); (L.W.); (G.J.); (A.H.-K.); (F.E.); (C.M.-T.); (M.S.)
| | - Anita Schmitt
- Department of Hematology, Oncology and Rheumatology, Heidelberg University Hospital, Im Neuenheimer Feld 410, 69120 Heidelberg, Germany; (A.R.); (A.K.); (A.K.); (B.N.); (D.S.); (L.W.); (G.J.); (A.H.-K.); (F.E.); (C.M.-T.); (M.S.)
- Correspondence: ; Tel.: +49-6221-56-6614; Fax: +49-6221-56-5740
| |
Collapse
|
17
|
Christodoulou I, Rahnama R, Ravich JW, Seo J, Zolov SN, Marple AN, Markovitz DM, Bonifant CL. Glycoprotein Targeted CAR-NK Cells for the Treatment of SARS-CoV-2 Infection. Front Immunol 2022; 12:763460. [PMID: 35003077 PMCID: PMC8732772 DOI: 10.3389/fimmu.2021.763460] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2021] [Accepted: 11/25/2021] [Indexed: 12/19/2022] Open
Abstract
H84T-Banana Lectin (BanLec) CAR-NK cells bind high mannose glycosites that decorate the SARS-CoV-2 envelope, thereby decreasing cellular infection in a model of SARS-CoV-2. H84T-BanLec CAR-NK cells are innate effector cells, activated by virus. This novel cellular agent is a promising therapeutic, capable of clearing circulating SARS-CoV-2 virus and infected cells. Banana Lectin (BanLec) binds high mannose glycans on viral envelopes, exerting an anti-viral effect. A point mutation (H84T) divorces BanLec mitogenicity from antiviral activity. SARS-CoV-2 contains high mannose glycosites in proximity to the receptor binding domain of the envelope Spike (S) protein. We designed a chimeric antigen receptor (CAR) that incorporates H84T-BanLec as the extracellular moiety. Our H84T-BanLec CAR was devised to specifically direct NK cell binding of SARS-CoV-2 envelope glycosites to promote viral clearance. The H84T-BanLec CAR was stably expressed at high density on primary human NK cells during two weeks of ex vivo expansion. H84T-BanLec CAR-NK cells reduced S-protein pseudotyped lentiviral infection of 293T cells expressing ACE2, the receptor for SARS-CoV-2. NK cells were activated to secrete inflammatory cytokines when in culture with virally infected cells. H84T-BanLec CAR-NK cells are a promising cell therapy for further testing against wild-type SARS-CoV-2 virus in models of SARS-CoV-2 infection. They may represent a viable off-the-shelf immunotherapy for patients suffering from COVID-19.
Collapse
Affiliation(s)
- Ilias Christodoulou
- The Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, MD, United States
| | - Ruyan Rahnama
- The Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, MD, United States.,Department of Pediatrics, Johns Hopkins University School of Medicine, Baltimore, MD, United States
| | - Jonas W Ravich
- The Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, MD, United States
| | - Jaesung Seo
- The Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, MD, United States
| | - Sergey N Zolov
- Department of Internal Medicine, University of Michigan, Ann Arbor, MI, United States
| | - Andrew N Marple
- The Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, MD, United States.,Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, MD, United States
| | - David M Markovitz
- Department of Internal Medicine, University of Michigan, Ann Arbor, MI, United States.,Division of Infectious Diseases, Department of Internal Medicine, and the Programs in Immunology, Cellular and Molecular Biology, and Cancer Biology, University of Michigan, Ann Arbor, MI, United States
| | - Challice L Bonifant
- The Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, MD, United States.,Department of Pediatrics, Johns Hopkins University School of Medicine, Baltimore, MD, United States
| |
Collapse
|
18
|
Alhomoud M, Martinet J, Sugita M, Gomez-Arteaga A, Guzman ML. Methods to monitor in vivo expansion and efficacy of CAR-T cells in preclinical models. Methods Cell Biol 2022; 167:185-201. [DOI: 10.1016/bs.mcb.2021.11.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
|
19
|
Sensitivity and Specificity of CD19.CAR-T Cell Detection by Flow Cytometry and PCR. Cells 2021; 10:cells10113208. [PMID: 34831430 PMCID: PMC8621201 DOI: 10.3390/cells10113208] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2021] [Revised: 11/07/2021] [Accepted: 11/10/2021] [Indexed: 11/16/2022] Open
Abstract
Chimeric-antigen-receptor-T (CAR-T) cells are currently revolutionizing the field of cancer immunotherapy. Therefore, there is an urgent need for CAR-T cell monitoring by clinicians to assess cell expansion and persistence in patients. CAR-T cell manufacturers and researchers need to evaluate transduction efficiency and vector copy number for quality control. Here, CAR expression was analyzed in peripheral blood samples from patients and healthy donors by flow cytometry with four commercially available detection reagents and on the gene level by quantitative polymerase chain reaction (qPCR). Flow cytometric analysis of CAR expression showed higher mean CAR expression values for CD19 CAR detection reagent and the F(ab')2 antibody than Protein L and CD19 Protein. In addition, the CD19 CAR detection reagent showed a significantly lower median background staining of 0.02% (range 0.007-0.06%) when compared to the F(ab')2 antibody, CD19 protein and Protein L with 0.80% (range 0.47-1.58%), 0.65% (range 0.25-1.35%) and 0.73% (range 0.44-1.23%). Furthermore, flow cytometry-based CAR-T cell frequencies by CD19 CAR detection reagent showed a good correlation with qPCR results. In conclusion, quality control of CAR-T cell products can be performed by FACS and qPCR. For the monitoring of CAR-T cell frequencies by FACS in patients, CAR detection reagents with a low background staining are preferable.
Collapse
|
20
|
Fatal late-onset CAR T-cell-mediated encephalitis after axicabtagene-ciloleucel in a patient with large B-cell lymphoma. Blood Adv 2021; 5:3789-3793. [PMID: 34496024 DOI: 10.1182/bloodadvances.2021004889] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2021] [Accepted: 06/02/2021] [Indexed: 11/20/2022] Open
Abstract
Treatment with CD19-directed (CAR) T cells has evolved as a standard of care for multiply relapsed or refractory large B-cell lymphoma (r/r LBCL). A common side effect of this treatment is the immune effector cell-associated neurotoxicity syndrome (ICANS). Severe ICANS can occur in up to 30% to 40% of patients treated with axicabtagene-ciloleucel (axi-cel), usually within the first 4 weeks after administration of the dose and usually responding well to steroids. We describe a case of progressive central neurotoxicity occurring 9 months after axi-cel infusion in a patient with r/r LBCL who had undergone a prior allogeneic hematopoietic cell transplant. Despite extensive systemic and intrathecal immunosuppression, neurological deterioration was inexorable and eventually fatal within 5 months. High CAR T-cell DNA copy numbers and elevated levels of interleukin-1 (IL-1) and IL-6 were found in the cerebral spinal fluid as clinical symptoms emerged, and CAR T-cell brain infiltration was observed on autopsy, suggesting that CAR T cells played a major pathogenetic role. This case of unexpected, devastating, late neurotoxicity warrants intensified investigation of neurological off-target effects of CD19-directed CAR T cells and highlights the need for continuous monitoring for late toxicities in this vulnerable patient population.
Collapse
|
21
|
Haderbache R, Warda W, Hervouet E, da Rocha MN, Trad R, Allain V, Nicod C, Thieblemeont C, Boissel N, Varlet P, Agha IY, Bouquet L, Guiot M, Venet F, Sujobert P, Roussel X, Rouzaire PO, Caillot D, Casasnovas O, Bories JC, Bachy E, Caillat-Zucman S, Deschamps M, Ferrand C. Droplet digital PCR allows vector copy number assessment and monitoring of experimental CAR T cells in murine xenograft models or approved CD19 CAR T cell-treated patients. J Transl Med 2021; 19:265. [PMID: 34154602 PMCID: PMC8215786 DOI: 10.1186/s12967-021-02925-z] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2021] [Accepted: 06/03/2021] [Indexed: 01/04/2023] Open
Abstract
BACKGROUND Genetically engineered chimeric antigen receptor (CAR) T lymphocytes are promising therapeutic tools for cancer. Four CAR T cell drugs, including tisagenlecleucel (tisa-cel) and axicabtagene-ciloleucel (axi-cel), all targeting CD19, are currently approved for treating B cell malignancies. Flow cytometry (FC) remains the standard for monitoring CAR T cells using a recombinant biotinylated target protein. Nevertheless, there is a need for additional tools, and the challenge is to develop an easy, relevant, highly sensitive, reproducible, and inexpensive detection method. Molecular tools can meet this need to specifically monitor long-term persistent CAR T cells. METHODS Based on 2 experimental CAR T cell constructs, IL-1RAP and CS1, we designed 2 quantitative digital droplet (ddPCR) PCR assays. By targeting the 4.1BB/CD3z (28BBz) or 28/CD3z (28z) junction area, we demonstrated that PCR assays can be applied to approved CD19 CAR T drugs. Both 28z and 28BBz ddPCR assays allow determination of the average vector copy number (VCN) per cell. We confirmed that the VCN is dependent on the multiplicity of infection and verified that the VCN of our experimental or GMP-like IL-1RAP CAR T cells met the requirement (< 5 VCN/cell) for delivery to the clinical department, similar to approved axi-cel or tisa-cel drugs. RESULTS 28BBz and 28z ddPCR assays applied to 2 tumoral (acute myeloid leukemia (AML) or multiple myeloma (MM) xenograft humanized NSG mouse models allowed us to quantify the early expansion (up to day 30) of CAR T cells after injection. Interestingly, following initial expansion, when circulating CAR T cells were challenged with the tumor, we noted a second expansion phase. Investigation of the bone marrow, spleen and lung showed that CAR T cells disseminated more within these tissues in mice previously injected with leukemic cell lines. Finally, circulating CAR T cell ddPCR monitoring of R/R acute lymphoid leukemia or diffuse large B cell lymphoma (n = 10 for tisa-cel and n = 7 for axi-cel) patients treated with both approved CAR T cells allowed detection of early expansion, which was highly correlated with FC, as well as long-term persistence (up to 450 days), while FC failed to detect these events. CONCLUSION Overall, we designed and validated 2 ddPCR assays allowing routine or preclinical monitoring of early- and long-term circulating approved or experimental CAR T cells, including our own IL-1RAP CAR T cells, which will be evaluated in an upcoming phase I clinical trial.
Collapse
Affiliation(s)
- Rafik Haderbache
- INSERM UMR1098, Right, EFSBFC, UFC, Laboratoire de Thérapeutique Immuno-Moléculaire Et Cellulaire Des Cancers, 8 rue du Dr Jean François Xavier Girod, 25020, Besançon, France
| | - Walid Warda
- INSERM UMR1098, Right, EFSBFC, UFC, Laboratoire de Thérapeutique Immuno-Moléculaire Et Cellulaire Des Cancers, 8 rue du Dr Jean François Xavier Girod, 25020, Besançon, France
| | - Eric Hervouet
- INSERM UMR1098, Right, EFSBFC, UFC, Laboratoire de Thérapeutique Immuno-Moléculaire Et Cellulaire Des Cancers, 8 rue du Dr Jean François Xavier Girod, 25020, Besançon, France
| | - Mathieu Neto da Rocha
- INSERM UMR1098, Right, EFSBFC, UFC, Laboratoire de Thérapeutique Immuno-Moléculaire Et Cellulaire Des Cancers, 8 rue du Dr Jean François Xavier Girod, 25020, Besançon, France
| | - Rim Trad
- INSERM UMR1098, Right, EFSBFC, UFC, Laboratoire de Thérapeutique Immuno-Moléculaire Et Cellulaire Des Cancers, 8 rue du Dr Jean François Xavier Girod, 25020, Besançon, France
| | - Vincent Allain
- Hôpital Saint-Louis, Assistance Publique-Hôpitaux de Paris (AP-HP), Université de Paris, Service d'Immunologie, Paris, France
| | - Clementine Nicod
- INSERM UMR1098, Right, EFSBFC, UFC, Laboratoire de Thérapeutique Immuno-Moléculaire Et Cellulaire Des Cancers, 8 rue du Dr Jean François Xavier Girod, 25020, Besançon, France
| | - Catherine Thieblemeont
- Hôpital Saint-Louis, Assistance Publique-Hôpitaux de Paris (AP-HP), Université de Paris, Service Hématologie, Paris, France
| | - Nicolas Boissel
- Hôpital Saint-Louis, Assistance Publique-Hôpitaux de Paris (AP-HP), Université de Paris, Service Hématologie, Paris, France
| | | | | | - Lucie Bouquet
- INSERM UMR1098, Right, EFSBFC, UFC, Laboratoire de Thérapeutique Immuno-Moléculaire Et Cellulaire Des Cancers, 8 rue du Dr Jean François Xavier Girod, 25020, Besançon, France
| | - Melanie Guiot
- INSERM UMR1098, Right, EFSBFC, UFC, Laboratoire de Thérapeutique Immuno-Moléculaire Et Cellulaire Des Cancers, 8 rue du Dr Jean François Xavier Girod, 25020, Besançon, France
| | - Fabienne Venet
- Hospices Civils de Lyon, Immunology Laboratory, Edouard Herriot Hospital, Lyon, France
| | - Pierre Sujobert
- Hospices Civils de Lyon, Hôpital Lyon Sud, Service d'Hématologie Biologique, Lyon, France
| | - Xavier Roussel
- INSERM UMR1098, Right, EFSBFC, UFC, Laboratoire de Thérapeutique Immuno-Moléculaire Et Cellulaire Des Cancers, 8 rue du Dr Jean François Xavier Girod, 25020, Besançon, France
| | - Paul-Oliver Rouzaire
- UFR de Pharmacie, EA CHELTER 7453, CHU de Clermont-Ferrand, Clermont-Ferrand, France
| | - Denis Caillot
- Hematology Clinical Department, Mitterrand Hospital, Dijon, France
| | | | | | - Emmanuel Bachy
- Hospices Civils de Lyon, Hospital Lyon Sud, Service d'Hématologie Clinique, Lyon, France
| | - Sophie Caillat-Zucman
- Hôpital Saint-Louis, Assistance Publique-Hôpitaux de Paris (AP-HP), Université de Paris, Service d'Immunologie, Paris, France
| | - Marina Deschamps
- INSERM UMR1098, Right, EFSBFC, UFC, Laboratoire de Thérapeutique Immuno-Moléculaire Et Cellulaire Des Cancers, 8 rue du Dr Jean François Xavier Girod, 25020, Besançon, France
| | - Christophe Ferrand
- INSERM UMR1098, Right, EFSBFC, UFC, Laboratoire de Thérapeutique Immuno-Moléculaire Et Cellulaire Des Cancers, 8 rue du Dr Jean François Xavier Girod, 25020, Besançon, France.
| |
Collapse
|
22
|
Laurent E, Sieber A, Salzer B, Wachernig A, Seigner J, Lehner M, Geyeregger R, Kratzer B, Jäger U, Kunert R, Pickl WF, Traxlmayr MW. Directed Evolution of Stabilized Monomeric CD19 for Monovalent CAR Interaction Studies and Monitoring of CAR-T Cell Patients. ACS Synth Biol 2021; 10:1184-1198. [PMID: 33843201 PMCID: PMC8155657 DOI: 10.1021/acssynbio.1c00010] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
CD19 is among the most relevant targets in cancer immunotherapy. However, its extracellular domain (ECD) is prone to aggregation and misfolding, representing a major obstacle for the development and analysis of CD19-targeted therapeutics. Here, we engineered stabilized CD19-ECD (termed SuperFolder) variants, which also showed improved expression rates and, in contrast to the wild type protein, they could be efficiently purified in their monomeric forms. Despite being considerably more stable, these engineered mutants largely preserved the wild type sequence (>98.8%). We demonstrate that the variant SF05 enabled the determination of the monovalent affinity between CD19 and a clinically approved FMC63-based CAR, as well as monitoring and phenotypic characterization of CD19-directed CAR-T cells in the blood of lymphoma patients. We anticipate that the SuperFolder mutants generated in this study will be highly valuable tools for a range of applications in basic immunology and CD19-targeted cancer immunotherapy.
Collapse
Affiliation(s)
- Elisabeth Laurent
- Department of Biotechnology and BOKU Core Facility Biomolecular and Cellular Analysis, BOKU - University of Natural Resources and Life Sciences, Muthgasse 18, 1190 Vienna, Austria
| | - Anna Sieber
- Department of Biotechnology, BOKU - University of Natural Resources and Life Sciences, Muthgasse 18, 1190 Vienna, Austria
| | - Benjamin Salzer
- St. Anna Children’s Cancer Research Institute, Zimmermannplatz 10, 1090 Vienna, Austria
| | - Anna Wachernig
- Department of Biotechnology, BOKU - University of Natural Resources and Life Sciences, Muthgasse 18, 1190 Vienna, Austria
| | - Jacqueline Seigner
- Department of Chemistry, BOKU - University of Natural Resources and Life Sciences, Muthgasse 18, 1190 Vienna, Austria
| | - Manfred Lehner
- St. Anna Children’s Cancer Research Institute, Zimmermannplatz 10, 1090 Vienna, Austria
| | - René Geyeregger
- St. Anna Children’s Cancer Research Institute, Zimmermannplatz 10, 1090 Vienna, Austria
| | - Bernhard Kratzer
- Institute of Immunology, Center for Pathophysiology, Infectiology and Immunology, Medical University of Vienna, Lazarettgasse 19, 1090 Vienna, Austria
| | - Ulrich Jäger
- Department of Internal Medicine, Division of Hematology and Hemostaseology, Medical University of Vienna, Währinger Gürtel 18-20, 1090 Vienna, Austria
| | - Renate Kunert
- Department of Biotechnology, BOKU - University of Natural Resources and Life Sciences, Muthgasse 18, 1190 Vienna, Austria
| | - Winfried F. Pickl
- Institute of Immunology, Center for Pathophysiology, Infectiology and Immunology, Medical University of Vienna, Lazarettgasse 19, 1090 Vienna, Austria
| | - Michael W. Traxlmayr
- Department of Chemistry, BOKU - University of Natural Resources and Life Sciences, Muthgasse 18, 1190 Vienna, Austria
| |
Collapse
|
23
|
Infection Complications after Lymphodepletion and Dosing of Chimeric Antigen Receptor T (CAR-T) Cell Therapy in Patients with Relapsed/Refractory Acute Lymphoblastic Leukemia or B Cell Non-Hodgkin Lymphoma. Cancers (Basel) 2021; 13:cancers13071684. [PMID: 33918340 PMCID: PMC8038233 DOI: 10.3390/cancers13071684] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2021] [Revised: 03/26/2021] [Accepted: 03/29/2021] [Indexed: 12/22/2022] Open
Abstract
Chimeric antigen receptor T (CAR-T) cell therapy has proven to be very effective in patients with relapsed/refractory acute lymphoblastic leukemia (ALL) and non-Hodgkin lymphoma (NHL). However, infections-related either due to lymphodepletion or the CAR-T cell therapy itself-can result in severe and potentially life-threatening complications, while side effects such as cytokine release syndrome (CRS) might complicate differential diagnosis. Sixty-seven dosings of CAR-T cells in sixty adult patients with NHL (85%) and ALL (15%) receiving CAR-T cell therapy were assessed for infectious complications. Almost two-thirds of patients (61%) developed fever following lymphodepletion and CAR-T cell dosing. Microbiological or radiological findings were observed in 25% of all cases (bacterial 12%, viral 5%, fungal 8%). Inpatient infections were associated with more lines of therapy and more severe CRS. However, overall serious complications were rare after CAR-T therapy, with one patient dying of infection. Pathogen detection after inpatient stay was infrequent and mostly occurred in the first 90 days after dosing. Infections in CAR-T cell treated patents are common. Fast and suitable identification and treatment are crucial in these heavily pretreated and immunocompromised patients. In most cases infectious complications are manageable. Nonetheless, standardized anti-infective prophylaxis and supportive therapy are mandatory to reduce morbidity and mortality in CAR-T cell therapy.
Collapse
|
24
|
Selim AG, Minson A, Blombery P, Dickinson M, Harrison SJ, Anderson MA. CAR-T cell therapy: practical guide to routine laboratory monitoring. Pathology 2021; 53:408-415. [PMID: 33685719 DOI: 10.1016/j.pathol.2021.02.002] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2021] [Revised: 02/08/2021] [Accepted: 02/09/2021] [Indexed: 01/24/2023]
Abstract
Chimeric antigen receptor (CAR)-T cell therapy is a genetically-modified cellular immunotherapy that has a current established role in the treatment of relapsed/refractory B-cell acute lymphoblastic leukaemia and diffuse large B-cell lymphoma, with emerging utility in a spectrum of other haematological and solid organ malignancies. It is associated with a number of characteristic toxicities, most notably cytokine release syndrome and neurotoxicity, for which laboratory testing can aid in the prediction of severity and in monitoring. Other toxicities, such as cytopenias/marrow hypoplasia, hypogammagloblinaemia and delayed immune reconstitution are recognised and require monitoring due to the implications for infection risk and prophylaxis. The detection or quantitation of circulating CAR-T can be clinically useful, and is achieved through both direct methods, if available, or indirect/surrogate methods. It is important that the laboratory is informed of the CAR-T therapy and target antigen whenever tissue is collected, both for response assessment and investigation of possible relapse, so that the expression of the relevant antigen can be assessed, in order to distinguish antigen-positive and -negative relapses. Finally, the measurement of circulating tumour DNA has an evolving role in the surveillance of malignancy, with evidence of its utility in the post-CAR-T setting, including predicting patients who will inevitably experience frank relapse, potentially allowing for pre-emptive therapy.
Collapse
Affiliation(s)
- Adrian G Selim
- Clinical Hematology, The Royal Melbourne Hospital and Peter MacCallum Cancer Centre, Melbourne, Vic, Australia; Centre of Excellence in Cellular Immunotherapy, Peter MacCallum Cancer Centre, Melbourne, Vic, Australia
| | - Adrian Minson
- Clinical Hematology, The Royal Melbourne Hospital and Peter MacCallum Cancer Centre, Melbourne, Vic, Australia
| | - Piers Blombery
- Clinical Hematology, The Royal Melbourne Hospital and Peter MacCallum Cancer Centre, Melbourne, Vic, Australia; Sir Peter MacCallum Department of Oncology, University of Melbourne, Parkville, Vic, Australia
| | - Michael Dickinson
- Clinical Hematology, The Royal Melbourne Hospital and Peter MacCallum Cancer Centre, Melbourne, Vic, Australia
| | - Simon J Harrison
- Clinical Hematology, The Royal Melbourne Hospital and Peter MacCallum Cancer Centre, Melbourne, Vic, Australia; Centre of Excellence in Cellular Immunotherapy, Peter MacCallum Cancer Centre, Melbourne, Vic, Australia; Sir Peter MacCallum Department of Oncology, University of Melbourne, Parkville, Vic, Australia
| | - Mary Ann Anderson
- Clinical Hematology, The Royal Melbourne Hospital and Peter MacCallum Cancer Centre, Melbourne, Vic, Australia; Centre of Excellence in Cellular Immunotherapy, Peter MacCallum Cancer Centre, Melbourne, Vic, Australia; Division of Blood Cells and Blood Cancer, The Walter and Eliza Hall Institute, Parkville, Vic, Australia.
| |
Collapse
|
25
|
Assessment of CAR T Cell Frequencies in Axicabtagene Ciloleucel and Tisagenlecleucel Patients Using Duplex Quantitative PCR. Cancers (Basel) 2020; 12:cancers12102820. [PMID: 33007926 PMCID: PMC7601213 DOI: 10.3390/cancers12102820] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2020] [Revised: 09/21/2020] [Accepted: 09/27/2020] [Indexed: 02/06/2023] Open
Abstract
Chimeric antigen receptor (CAR) T cell (CART) therapy has been established as a treatment option for patients with CD19-positive lymphoid malignancies in both the refractory and the relapsed setting. Displaying significant responses in clinical trials, two second-generation CART products directed against CD19, axicabtagene ciloleucel (axi-cel) and tisagenlecleucel (tisa-cel), have been approved and integrated into the clinical routine. However, experimental assay for quantitative monitoring of both of these CART products in treated patients in the open domain are lacking. To address this issue, we established and validated a quantitative single copy gene (SCG)-based duplex (DP)-PCR assay (SCG-DP-PCR) to quantify CARTs based on the FMC63 single chain variable fragment (scFv), i.e., axi-cel and tisa-cel. This quantitative PCR (qPCR) approach operates without standard curves or calibrator samples, offers a tool to assess cellular kinetics of FMC63 CARTs and allows direct comparison of CART-copies in axi-cel versus tisa-cel patient samples. For treating physicians, SCG-DP-PCR is an important tool to monitor CARTs and guide clinical decisions regarding CART effects in respective patients.
Collapse
|
26
|
Abstract
Chimeric antigen receptor-T (CAR-T) cell therapy is a promising frontier of immunoengineering and cancer immunotherapy. Methods that detect, quantify, track, and visualize the CAR, have catalyzed the rapid advancement of CAR-T cell therapy from preclinical models to clinical adoption. For instance, CAR-staining/labeling agents have enabled flow cytometry analysis, imaging applications, cell sorting, and high-dimensional clinical profiling. Molecular assays, such as quantitative polymerase chain reaction, integration site analysis, and RNA-sequencing, have characterized CAR transduction, expression, and in vivo CAR-T cell expansion kinetics. In vitro visualization methods, including confocal and total internal reflection fluorescence microscopy, have captured the molecular details underlying CAR immunological synapse formation, signaling, and cytotoxicity. In vivo tracking methods, including two-photon microscopy, bioluminescence imaging, and positron emission tomography scanning, have monitored CAR-T cell biodistribution across blood, tissue, and tumor. Here, we review the plethora of CAR detection methods, which can operate at the genomic, transcriptomic, proteomic, and organismal levels. For each method, we discuss: (1) what it measures; (2) how it works; (3) its scientific and clinical importance; (4) relevant examples of its use; (5) specific protocols for CAR detection; and (6) its strengths and weaknesses. Finally, we consider current scientific and clinical needs in order to provide future perspectives for improved CAR detection.
Collapse
Affiliation(s)
- Yifei Hu
- Pritzker School of Molecular Engineering, University of Chicago, Chicago, IL, United States
- Pritzker School of Medicine, University of Chicago, Chicago, IL, United States
| | - Jun Huang
- Pritzker School of Molecular Engineering, University of Chicago, Chicago, IL, United States
| |
Collapse
|
27
|
Accurate In-Vivo Quantification of CD19 CAR-T Cells after Treatment with Axicabtagene Ciloleucel (Axi-Cel) and Tisagenlecleucel (Tisa-Cel) Using Digital PCR. Cancers (Basel) 2020; 12:cancers12071970. [PMID: 32698364 PMCID: PMC7409173 DOI: 10.3390/cancers12071970] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2020] [Revised: 07/14/2020] [Accepted: 07/17/2020] [Indexed: 01/01/2023] Open
Abstract
Immunotherapy with CD19-specific chimeric antigen receptor (CAR-) T cells has shown excellent efficacy in relapsed/refractory B-cell cancers. The in vivo expansion and persistence of CAR-T cells after infusion are important response- and toxicity-determining variables, but diagnostic tools are largely missing. We showed previously for axi-cel that digital PCR (dPCR) is excellently suited to monitoring CAR-T cells in vivo. Here, we aimed to develop an analogous dPCR assay for tisa-cel. To do so, we cloned and sequenced the CAR construct from the lentiviral tisa-cel vector and designed primers and Black hole quencher (BHQ) probes complimentary to sequences present in the FMC63 scFv part of axi-cel (assay A), tisa-cel (T), and both constructs (U = “universal”). In conjunction with excellent specificity, all assays have a detection limit of one single CAR copy, corresponding to a sensitivity of approximately 1 in 5000 cells (0.02%) for 100 ng genomic DNA (for one vector copy per transduced cell). The new universal assay was first validated using patient samples previously quantified with the axi-cel-specific dPCR and thereafter applied to quantify and monitor adoptively transferred axi-cel and tisa-cel T cells in post-infusion samples (peripheral blood, bone marrow, liquor, and ascites). Actual CAR-T counts per µl were calculated, taking into account vector copy and peripheral blood mononuclear cell (PBMC) numbers, and showed very good correlation with flow cytometry results. We conclude that our novel dPCR assay is optimally suited to monitoring tisa-cel and axi-cel CAR-T cells in real-time in various body fluids.
Collapse
|
28
|
Schubert ML, Dietrich S, Stilgenbauer S, Schmitt A, Pavel P, Kunz A, Bondong A, Wegner M, Stadtherr P, Jung S, Ho AD, Müller-Tidow C, Schmitt M, Dreger P. Feasibility and Safety of CD19 Chimeric Antigen Receptor T Cell Treatment for B Cell Lymphoma Relapse after Allogeneic Hematopoietic Stem Cell Transplantation. Biol Blood Marrow Transplant 2020; 26:1575-1580. [PMID: 32422254 DOI: 10.1016/j.bbmt.2020.04.025] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2020] [Revised: 04/20/2020] [Accepted: 04/23/2020] [Indexed: 12/21/2022]
Abstract
Although CD19-directed chimeric antigen receptor (CAR) T cells have been successfully used after a preceding allogeneic stem cell transplant (alloHCT) in patients with acute lymphoblastic leukemia, little is known about the feasibility and outcome of CAR T cell treatment in patients who have been previously allotransplanted for lymphoma. In a single-center retrospective analysis, course and outcome of all allografted patients treated with CD19 CAR constructs for B cell lymphoma between October 2018 and November 2019 were studied. CAR therapy consisted either of a third-generation CAR (HD-CAR-1) or of commercially manufactured axicabtagene ciloleucel (axi-cel; Gilead, Santa Monica, U.S.). Altogether, 10 CAR T cell dosings using recipient leukapheresis products were performed in 8 patients: 4 patients (2 mantle cell lymphoma, 2 chronic lymphocytic leukemia) received 6 dosings with HD-CAR-1 and 4 patients (all with diffuse large B cell lymphoma) received 4 dosings with axi-cel. Overall, 6 of 8 patients (75%) responded. CAR treatment was well tolerated with grade ≥ 3 cytokine release syndrome and neurotoxicity each being observed after 1 of 10 dosings. A single patient had moderate chronic graft-versus-host disease. Of note, 3 of 4 patients who received axi-cel had ongoing grade ≥ 3 cytopenia 3 months postdosing, whereas prolonged cytopenia was not observed in 9 alloHCT-naive patients who received axi-cel during the same time period. In conclusion, CAR T cell treatment from recipient-derived leukapheresis products after a prior alloHCT appears to be feasible, effective, and safe in patients with B cell lymphoma. Protracted cytopenia after axi-cel treatment is a matter of concern and requires further exploration.
Collapse
Affiliation(s)
- Maria-Luisa Schubert
- Department of Internal Medicine V, University of Heidelberg, Heidelberg, Germany.
| | - Sascha Dietrich
- Department of Internal Medicine V, University of Heidelberg, Heidelberg, Germany; German Cancer Consortium (DKTK) and German Cancer Research Center (DKFZ)/National Centre for Tumor Diseases (NCT), Heidelberg, Germany
| | - Stephan Stilgenbauer
- Department of Internal Medicine III, Ulm University Medical Center, Ulm, Germany.; Department of Internal Medicine I, Saarland University Medical Center, Homburg, Germany
| | - Anita Schmitt
- Department of Internal Medicine V, University of Heidelberg, Heidelberg, Germany
| | - Petra Pavel
- Stem Cell Laboratory, Institute for Clinical Transfusion Medicine and Cell Therapy (IKTZ), German Red Cross Blood Service Baden-Württemberg-Hessen, Heidelberg, Germany
| | - Alexander Kunz
- Department of Internal Medicine V, University of Heidelberg, Heidelberg, Germany
| | - Andrea Bondong
- Department of Internal Medicine V, University of Heidelberg, Heidelberg, Germany
| | - Mandy Wegner
- Department of Internal Medicine V, University of Heidelberg, Heidelberg, Germany
| | - Peter Stadtherr
- Department of Internal Medicine V, University of Heidelberg, Heidelberg, Germany
| | - Susanne Jung
- Diakonissenkrankenhaus und Paulinenhilfe gGmbH, Diakonie-Klinikum, Stuttgart, Germany
| | - Anthony D Ho
- Department of Internal Medicine V, University of Heidelberg, Heidelberg, Germany; German Cancer Consortium (DKTK) and German Cancer Research Center (DKFZ)/National Centre for Tumor Diseases (NCT), Heidelberg, Germany
| | - Carsten Müller-Tidow
- Department of Internal Medicine V, University of Heidelberg, Heidelberg, Germany; German Cancer Consortium (DKTK) and German Cancer Research Center (DKFZ)/National Centre for Tumor Diseases (NCT), Heidelberg, Germany
| | - Michael Schmitt
- Department of Internal Medicine V, University of Heidelberg, Heidelberg, Germany; German Cancer Consortium (DKTK) and German Cancer Research Center (DKFZ)/National Centre for Tumor Diseases (NCT), Heidelberg, Germany
| | - Peter Dreger
- Department of Internal Medicine V, University of Heidelberg, Heidelberg, Germany; German Cancer Consortium (DKTK) and German Cancer Research Center (DKFZ)/National Centre for Tumor Diseases (NCT), Heidelberg, Germany
| |
Collapse
|