1
|
Knelangen N, Bader U, Maniaki E, Langan PS, Engert F, Drees B, Schwarzer J, Kotter B, Kiefer L, Gattinoni L, Engels B, Mittelstaet J, Webster B. CAR T cells re-directed by a rationally designed human peptide tag demonstrate efficacy in preclinical models. Cytotherapy 2025:S1465-3249(25)00592-4. [PMID: 40257412 DOI: 10.1016/j.jcyt.2025.03.506] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2025] [Revised: 03/15/2025] [Accepted: 03/17/2025] [Indexed: 04/22/2025]
Abstract
Currently, only a few chimeric antigen receptor (CAR) T cell therapies have been approved by the Food and Drug Administration and European Medicines Agency for the treatment of B-cell malignancies. To enable broader application of the CAR T cell technology in other indications, improved control and flexible targeting of multiple tumor antigens are required. Here, we developed a novel adapter CAR (AdCAR) T cell platform for flexible targeting of multiple tumor antigens. This platform is based on a short peptide tag derived from an interdomain region of fibroblast growth factor receptor 2 (FGFR2), commonly mutated in cancer. To select AdCARs specific for mutated FGFR2-derived peptide tags, a multistep pooled screening approach in primary T cells was employed, incorporating MACS separation and next-generation sequencing. The resulting AdCAR was highly specific for the FGFR2-derived peptide tag. Using different in vitro and in vivo model systems, the activity of AdCAR T cells was shown to be strictly dependent on the presence of the adapter and corresponding target antigen. Moreover, AdCAR T cells could be redirected to different target antigens by the addition of respective adapter molecules (AM). Finally, in situ expression of functional AM in primary T cells under control of a drug-inducible promoter system was demonstrated, highlighting the potential for controlling the activity of AdCAR T cells by cellular micropharmacies.
Collapse
Affiliation(s)
- Nele Knelangen
- Research and Development, Miltenyi Biotec, Bergisch Gladbach, Germany; Division of Functional Immune Cell Modulation, Leibniz Institute for Immunotherapy, Regensburg, Germany.
| | - Ulrika Bader
- Research and Development, Miltenyi Biotec, Bergisch Gladbach, Germany.
| | - Evangelia Maniaki
- Research and Development, Miltenyi Biotec, Bergisch Gladbach, Germany
| | - Patricia S Langan
- Research and Development, Lentigen Technology Inc., A Miltenyi Biotec Company, Gaithersburg, Maryland, USA
| | - Fabian Engert
- Research and Development, Miltenyi Biotec, Bergisch Gladbach, Germany
| | - Britta Drees
- Research and Development, Miltenyi Biotec, Bergisch Gladbach, Germany
| | - Juliane Schwarzer
- Research and Development, Miltenyi Biotec, Bergisch Gladbach, Germany
| | - Bettina Kotter
- Research and Development, Miltenyi Biotec, Bergisch Gladbach, Germany
| | - Lukas Kiefer
- Research and Development, Miltenyi Biotec, Bergisch Gladbach, Germany
| | - Luca Gattinoni
- Division of Functional Immune Cell Modulation, Leibniz Institute for Immunotherapy, Regensburg, Germany; University of Regensburg, Regensburg, Germany
| | - Boris Engels
- Research and Development, Miltenyi Biotec, Bergisch Gladbach, Germany
| | - Joerg Mittelstaet
- Faculty of Life Sciences, Reutlingen University, Reutlingen, Germany
| | | |
Collapse
|
2
|
Kleinboehl EW, Laoharawee K, Jensen JD, Peterson JJ, Slipek NJ, Wick BJ, Johnson MJ, Webber BR, Moriarity BS. Engineering memory T cells as a platform for long-term enzyme replacement therapy in lysosomal storage disorders. Mol Ther 2024; 32:3865-3878. [PMID: 39367605 PMCID: PMC11573576 DOI: 10.1016/j.ymthe.2024.09.033] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2024] [Revised: 08/19/2024] [Accepted: 09/30/2024] [Indexed: 10/06/2024] Open
Abstract
Enzymopathy disorders are the result of missing or defective enzymes. Among these enzymopathies, mucopolysaccharidosis type I is a rare genetic lysosomal storage disorder caused by mutations in the gene encoding alpha-L-iduronidase (IDUA), which ultimately causes toxic buildup of glycosaminoglycans (GAGs). There is currently no cure and standard treatments provide insufficient relief to the skeletal structure and central nervous system (CNS). Human memory T (Tm) cells migrate throughout the body's tissues and can persist for years, making them an attractive approach for cellular-based, systemic enzyme replacement therapy. Here, we tested genetically engineered, IDUA-expressing Tm cells as a cellular therapy in an immunodeficient mouse model of MPS I. Our results demonstrate that a single dose of engineered Tm cells leads to detectable IDUA enzyme levels in the blood for up to 22 weeks and reduced urinary GAG excretion. Furthermore, engineered Tm cells take up residence in nearly all tested tissues, producing IDUA and leading to metabolic correction of GAG levels in the heart, lung, liver, spleen, kidney, bone marrow, and the CNS, although only minimal improved cognition was observed. Our study indicates that genetically engineered Tm cells hold great promise as a platform for cellular-based enzyme replacement therapy for the treatment of mucopolysaccharidosis type I and potentially many other enzymopathies and protein deficiencies.
Collapse
Affiliation(s)
- Evan W Kleinboehl
- Department of Pediatrics, University of Minnesota, Minneapolis, MN 55454, USA; Masonic Cancer Center, University of Minnesota, Minneapolis, MN 55455, USA; Center for Genome Engineering, University of Minnesota, Minneapolis, MN 55455, USA
| | - Kanut Laoharawee
- Department of Pediatrics, University of Minnesota, Minneapolis, MN 55454, USA; Masonic Cancer Center, University of Minnesota, Minneapolis, MN 55455, USA; Center for Genome Engineering, University of Minnesota, Minneapolis, MN 55455, USA
| | - Jacob D Jensen
- Department of Pediatrics, University of Minnesota, Minneapolis, MN 55454, USA; Masonic Cancer Center, University of Minnesota, Minneapolis, MN 55455, USA; Center for Genome Engineering, University of Minnesota, Minneapolis, MN 55455, USA
| | - Joseph J Peterson
- Department of Pediatrics, University of Minnesota, Minneapolis, MN 55454, USA; Masonic Cancer Center, University of Minnesota, Minneapolis, MN 55455, USA; Center for Genome Engineering, University of Minnesota, Minneapolis, MN 55455, USA
| | - Nicholas J Slipek
- Department of Pediatrics, University of Minnesota, Minneapolis, MN 55454, USA; Masonic Cancer Center, University of Minnesota, Minneapolis, MN 55455, USA; Center for Genome Engineering, University of Minnesota, Minneapolis, MN 55455, USA
| | - Bryce J Wick
- Department of Pediatrics, University of Minnesota, Minneapolis, MN 55454, USA; Masonic Cancer Center, University of Minnesota, Minneapolis, MN 55455, USA; Center for Genome Engineering, University of Minnesota, Minneapolis, MN 55455, USA
| | - Matthew J Johnson
- Department of Pediatrics, University of Minnesota, Minneapolis, MN 55454, USA; Masonic Cancer Center, University of Minnesota, Minneapolis, MN 55455, USA; Center for Genome Engineering, University of Minnesota, Minneapolis, MN 55455, USA
| | - Beau R Webber
- Department of Pediatrics, University of Minnesota, Minneapolis, MN 55454, USA; Masonic Cancer Center, University of Minnesota, Minneapolis, MN 55455, USA; Center for Genome Engineering, University of Minnesota, Minneapolis, MN 55455, USA.
| | - Branden S Moriarity
- Department of Pediatrics, University of Minnesota, Minneapolis, MN 55454, USA; Masonic Cancer Center, University of Minnesota, Minneapolis, MN 55455, USA; Center for Genome Engineering, University of Minnesota, Minneapolis, MN 55455, USA.
| |
Collapse
|
3
|
Skeate JG, Pomeroy EJ, Slipek NJ, Jones BJ, Wick BJ, Chang JW, Lahr WS, Stelljes EM, Patrinostro X, Barnes B, Zarecki T, Krueger JB, Bridge JE, Robbins GM, McCormick MD, Leerar JR, Wenzel KT, Hornberger KM, Walker K, Smedley D, Largaespada DA, Otto N, Webber BR, Moriarity BS. Evolution of the clinical-stage hyperactive TcBuster transposase as a platform for robust non-viral production of adoptive cellular therapies. Mol Ther 2024; 32:1817-1834. [PMID: 38627969 PMCID: PMC11184336 DOI: 10.1016/j.ymthe.2024.04.024] [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: 08/31/2023] [Revised: 03/06/2024] [Accepted: 04/12/2024] [Indexed: 06/09/2024] Open
Abstract
Cellular therapies for the treatment of human diseases, such as chimeric antigen receptor (CAR) T and natural killer (NK) cells have shown remarkable clinical efficacy in treating hematological malignancies; however, current methods mainly utilize viral vectors that are limited by their cargo size capacities, high cost, and long timelines for production of clinical reagent. Delivery of genetic cargo via DNA transposon engineering is a more timely and cost-effective approach, yet has been held back by less efficient integration rates. Here, we report the development of a novel hyperactive TcBuster (TcB-M) transposase engineered through structure-guided and in vitro evolution approaches that achieves high-efficiency integration of large, multicistronic CAR-expression cassettes in primary human cells. Our proof-of-principle TcB-M engineering of CAR-NK and CAR-T cells shows low integrated vector copy number, a safe insertion site profile, robust in vitro function, and improves survival in a Burkitt lymphoma xenograft model in vivo. Overall, TcB-M is a versatile, safe, efficient and open-source option for the rapid manufacture and preclinical testing of primary human immune cell therapies through delivery of multicistronic large cargo via transposition.
Collapse
Affiliation(s)
- Joseph G Skeate
- Department of Pediatrics, University of Minnesota, Minneapolis, MN 55455, USA; Masonic Cancer Center, University of Minnesota, Minneapolis, MN 55455, USA; Center for Genome Engineering, University of Minnesota, Minneapolis, MN 55455, USA
| | - Emily J Pomeroy
- Department of Pediatrics, University of Minnesota, Minneapolis, MN 55455, USA; Masonic Cancer Center, University of Minnesota, Minneapolis, MN 55455, USA; Center for Genome Engineering, University of Minnesota, Minneapolis, MN 55455, USA
| | - Nicholas J Slipek
- Department of Pediatrics, University of Minnesota, Minneapolis, MN 55455, USA; Masonic Cancer Center, University of Minnesota, Minneapolis, MN 55455, USA; Center for Genome Engineering, University of Minnesota, Minneapolis, MN 55455, USA
| | | | - Bryce J Wick
- Department of Pediatrics, University of Minnesota, Minneapolis, MN 55455, USA; Masonic Cancer Center, University of Minnesota, Minneapolis, MN 55455, USA; Center for Genome Engineering, University of Minnesota, Minneapolis, MN 55455, USA
| | - Jae-Woong Chang
- Department of Pediatrics, University of Minnesota, Minneapolis, MN 55455, USA; Masonic Cancer Center, University of Minnesota, Minneapolis, MN 55455, USA; Center for Genome Engineering, University of Minnesota, Minneapolis, MN 55455, USA
| | - Walker S Lahr
- Department of Pediatrics, University of Minnesota, Minneapolis, MN 55455, USA; Masonic Cancer Center, University of Minnesota, Minneapolis, MN 55455, USA; Center for Genome Engineering, University of Minnesota, Minneapolis, MN 55455, USA
| | - Erin M Stelljes
- Department of Pediatrics, University of Minnesota, Minneapolis, MN 55455, USA; Masonic Cancer Center, University of Minnesota, Minneapolis, MN 55455, USA; Center for Genome Engineering, University of Minnesota, Minneapolis, MN 55455, USA
| | | | | | | | - Joshua B Krueger
- Department of Pediatrics, University of Minnesota, Minneapolis, MN 55455, USA; Masonic Cancer Center, University of Minnesota, Minneapolis, MN 55455, USA; Center for Genome Engineering, University of Minnesota, Minneapolis, MN 55455, USA
| | - Jacob E Bridge
- Department of Pediatrics, University of Minnesota, Minneapolis, MN 55455, USA; Masonic Cancer Center, University of Minnesota, Minneapolis, MN 55455, USA; Center for Genome Engineering, University of Minnesota, Minneapolis, MN 55455, USA
| | - Gabrielle M Robbins
- Department of Pediatrics, University of Minnesota, Minneapolis, MN 55455, USA; Masonic Cancer Center, University of Minnesota, Minneapolis, MN 55455, USA; Center for Genome Engineering, University of Minnesota, Minneapolis, MN 55455, USA
| | - Madeline D McCormick
- Department of Pediatrics, University of Minnesota, Minneapolis, MN 55455, USA; Masonic Cancer Center, University of Minnesota, Minneapolis, MN 55455, USA; Center for Genome Engineering, University of Minnesota, Minneapolis, MN 55455, USA
| | | | | | | | | | | | - David A Largaespada
- Department of Pediatrics, University of Minnesota, Minneapolis, MN 55455, USA; Masonic Cancer Center, University of Minnesota, Minneapolis, MN 55455, USA; Center for Genome Engineering, University of Minnesota, Minneapolis, MN 55455, USA
| | - Neil Otto
- Bio-Techne, Minneapolis, MN 55413, USA
| | - Beau R Webber
- Department of Pediatrics, University of Minnesota, Minneapolis, MN 55455, USA; Masonic Cancer Center, University of Minnesota, Minneapolis, MN 55455, USA; Center for Genome Engineering, University of Minnesota, Minneapolis, MN 55455, USA.
| | - Branden S Moriarity
- Department of Pediatrics, University of Minnesota, Minneapolis, MN 55455, USA; Masonic Cancer Center, University of Minnesota, Minneapolis, MN 55455, USA; Center for Genome Engineering, University of Minnesota, Minneapolis, MN 55455, USA.
| |
Collapse
|
4
|
Laoharawee K, Kleinboehl EW, Jensen JD, Peterson JJ, Slipek NJ, Wick BJ, Johnson MJ, Webber BR, Moriarity BS. Engineering Memory T Cells as a platform for Long-Term Enzyme Replacement Therapy in Lysosomal Storage Disorders. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.04.23.590790. [PMID: 38712248 PMCID: PMC11071424 DOI: 10.1101/2024.04.23.590790] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/08/2024]
Abstract
Enzymopathy disorders are the result of missing or defective enzymes. Amongst these enzymopathies, mucopolysaccharidosis type I, is a rare genetic lysosomal storage disorder caused by mutations in the gene encoding alpha-L-iduronidase (IDUA), ultimately causes toxic build-up of glycosaminoglycans (GAGs). There is currently no cure and standard treatments provide insufficient relief to the skeletal structure and central nervous system (CNS). Human memory T cells (Tm) migrate throughout the body's tissues and can persist for years, making them an attractive approach for cellular-based, systemic enzyme replacement therapy. Here, we tested genetically engineered, IDUA-expressing Tm as a cellular therapy in an immunodeficient mouse model of MPS I. Our results demonstrate that a single dose of engineered Tm leads to detectable IDUA enzyme levels in the blood for up to 22 weeks and reduced urinary GAG excretion. Furthermore, engineered Tm take up residence in nearly all tested tissues, producing IDUA and leading to metabolic correction of GAG levels in the heart, lung, liver, spleen, kidney, bone marrow, and the CNS. Our study indicates that genetically engineered Tm holds great promise as a platform for cellular-based enzyme replacement therapy for the treatment of mucopolysaccharidosis type I and potentially many other enzymopathies and protein deficiencies.
Collapse
|
5
|
Madden MZ, Ye X, Chi C, Fisher EL, Wolf MM, Needle GA, Bader JE, Patterson AR, Reinfeld BI, Landis MD, Hathaway ES, Muka JE, O’Neil RT, Karijolich J, Philip M, Rathmell JC. Differential Effects of Glutamine Inhibition Strategies on Antitumor CD8 T Cells. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2023; 211:563-575. [PMID: 37341499 PMCID: PMC10526752 DOI: 10.4049/jimmunol.2200715] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/26/2022] [Accepted: 06/02/2023] [Indexed: 06/22/2023]
Abstract
Activated T cells undergo metabolic reprogramming to meet anabolic, differentiation, and functional demands. Glutamine supports many processes in activated T cells, and inhibition of glutamine metabolism alters T cell function in autoimmune disease and cancer. Multiple glutamine-targeting molecules are under investigation, yet the precise mechanisms of glutamine-dependent CD8 T cell differentiation remain unclear. We show that distinct strategies of glutamine inhibition by glutaminase-specific inhibition with small molecule CB-839, pan-glutamine inhibition with 6-diazo-5-oxo-l-norleucine (DON), or by glutamine-depleted conditions (No Q) produce distinct metabolic differentiation trajectories in murine CD8 T cells. T cell activation with CB-839 treatment had a milder effect than did DON or No Q treatment. A key difference was that CB-839-treated cells compensated with increased glycolytic metabolism, whereas DON and No Q-treated cells increased oxidative metabolism. However, all glutamine treatment strategies elevated CD8 T cell dependence on glucose metabolism, and No Q treatment caused adaptation toward reduced glutamine dependence. DON treatment reduced histone modifications and numbers of persisting cells in adoptive transfer studies, but those T cells that remained could expand normally upon secondary Ag encounter. In contrast, No Q-treated cells persisted well yet demonstrated decreased secondary expansion. Consistent with reduced persistence, CD8 T cells activated in the presence of DON had reduced ability to control tumor growth and reduced tumor infiltration in adoptive cell therapy. Overall, each approach to inhibit glutamine metabolism confers distinct effects on CD8 T cells and highlights that targeting the same pathway in different ways can elicit opposing metabolic and functional outcomes.
Collapse
Affiliation(s)
- Matthew Z. Madden
- Department of Pathology, Microbiology, and Immunology, Vanderbilt University Medical Center, Nashville, TN
| | - Xiang Ye
- Department of Pathology, Microbiology, and Immunology, Vanderbilt University Medical Center, Nashville, TN
| | - Channing Chi
- Department of Pathology, Microbiology, and Immunology, Vanderbilt University Medical Center, Nashville, TN
| | - Emilie L. Fisher
- Department of Pathology, Microbiology, and Immunology, Vanderbilt University Medical Center, Nashville, TN
| | - Melissa M. Wolf
- Department of Medicine, Vanderbilt University Medical Center, Nashville, TN
| | | | - Jackie E. Bader
- Department of Pathology, Microbiology, and Immunology, Vanderbilt University Medical Center, Nashville, TN
| | - Andrew R. Patterson
- Department of Pathology, Microbiology, and Immunology, Vanderbilt University Medical Center, Nashville, TN
| | | | - Madelyn D. Landis
- Department of Medicine, Vanderbilt University Medical Center, Nashville, TN
| | - Emma S. Hathaway
- Department of Pathology, Microbiology, and Immunology, Vanderbilt University Medical Center, Nashville, TN
| | - Jason E. Muka
- Department of Pathology, Microbiology, and Immunology, Vanderbilt University Medical Center, Nashville, TN
| | - Richard T. O’Neil
- Department of Veterans Affairs, Ralph H Johnson VA Medical Center and Department of Microbiology and Immunology, Medical University of South Carolina, Charleston, SC
| | - John Karijolich
- Department of Pathology, Microbiology, and Immunology, Vanderbilt University Medical Center, Nashville, TN
- Vanderbilt Center for Immunobiology, Nashville, TN
| | - Mary Philip
- Department of Medicine, Vanderbilt University Medical Center, Nashville, TN
- Vanderbilt Center for Immunobiology, Nashville, TN
| | - Jeffrey C. Rathmell
- Department of Pathology, Microbiology, and Immunology, Vanderbilt University Medical Center, Nashville, TN
- Vanderbilt Center for Immunobiology, Nashville, TN
| |
Collapse
|
6
|
Peek JL, Wilson MH. Cell and gene therapy for kidney disease. Nat Rev Nephrol 2023:10.1038/s41581-023-00702-3. [PMID: 36973494 DOI: 10.1038/s41581-023-00702-3] [Citation(s) in RCA: 24] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/07/2023] [Indexed: 03/29/2023]
Abstract
Kidney disease is a leading cause of morbidity and mortality across the globe. Current interventions for kidney disease include dialysis and renal transplantation, which have limited efficacy or availability and are often associated with complications such as cardiovascular disease and immunosuppression. There is therefore a pressing need for novel therapies for kidney disease. Notably, as many as 30% of kidney disease cases are caused by monogenic disease and are thus potentially amenable to genetic medicine, such as cell and gene therapy. Systemic disease that affects the kidney, such as diabetes and hypertension, might also be targetable by cell and gene therapy. However, although there are now several approved gene and cell therapies for inherited diseases that affect other organs, none targets the kidney. Promising recent advances in cell and gene therapy have been made, including in the kidney research field, suggesting that this form of therapy might represent a potential solution for kidney disease in the future. In this Review, we describe the potential for cell and gene therapy in treating kidney disease, focusing on recent genetic studies, key advances and emerging technologies, and we describe several crucial considerations for renal genetic and cell therapies.
Collapse
Affiliation(s)
- Jennifer L Peek
- Medical Scientist Training Program, Vanderbilt University School of Medicine, Nashville, TN, USA
- Department of Cell and Developmental Biology, Vanderbilt University, Nashville, TN, USA
| | - Matthew H Wilson
- Department of Cell and Developmental Biology, Vanderbilt University, Nashville, TN, USA.
- Department of Pharmacology, Vanderbilt University, Nashville, TN, USA.
- Department of Medicine, Division of Nephrology and Hypertension, Vanderbilt University Medical Center, Nashville, TN, USA.
- Department of Veterans Affairs, Tennessee Valley Health Services, Nashville, TN, USA.
| |
Collapse
|
7
|
CXCR4 and anti-BCMA CAR co-modified natural killer cells suppress multiple myeloma progression in a xenograft mouse model. Cancer Gene Ther 2022; 29:475-483. [PMID: 34471234 DOI: 10.1038/s41417-021-00365-x] [Citation(s) in RCA: 53] [Impact Index Per Article: 17.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2020] [Revised: 05/27/2021] [Accepted: 06/22/2021] [Indexed: 02/02/2023]
Abstract
The highly restricted expression of B-cell maturation antigen (BCMA) on plasma cells makes it an ideal target for chimeric antigen receptor (CAR) immune cell therapy against multiple myeloma (MM), a bone marrow cancer. To improve the infiltration of ex vivo expanded human natural killer (NK) cells into the bone marrow, we electroporated these cells with mRNA encoding the chemokine receptor CXCR4. The CXCR4-modified NK cells displayed increased in vitro migration toward the bone marrow niche-expressing chemokine CXCL12/SDF-1α and augmented infiltration into the bone marrow compartments in mice. We further modified the CXCR4-NK cells by electroporation of mRNA encoding a CAR targeting BCMA. After the intravenous injection of the double-modified NK cells into a xenograft mouse model of MM, we observed significantly reduced tumor burden in the femur region of the living mice and the extended survival of the tumor-bearing mice. Collectively, this study provides the experimental evidence that the co-expression of CXCR4 and anti-BCMA CAR on NK cells is a possible effective way to control MM progression.
Collapse
|
8
|
Nagree MS, Felizardo TC, Faber ML, Rybova J, Rupar CA, Foley SR, Fuller M, Fowler DH, Medin JA. Autologous, lentivirus-modified, T-rapa cell "micropharmacies" for lysosomal storage disorders. EMBO Mol Med 2022; 14:e14297. [PMID: 35298086 PMCID: PMC8988206 DOI: 10.15252/emmm.202114297] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2021] [Revised: 02/07/2022] [Accepted: 02/09/2022] [Indexed: 11/11/2022] Open
Abstract
T cells are the current choice for many cell therapy applications. They are relatively easy to access, expand in culture, and genetically modify. Rapamycin-conditioning ex vivo reprograms T cells, increasing their memory properties and capacity for survival, while reducing inflammatory potential and the amount of preparative conditioning required for engraftment. Rapamycin-conditioned T cells have been tested in patients and deemed to be safe to administer in numerous settings, with reduced occurrence of infusion-related adverse events. We demonstrate that ex vivo lentivirus-modified, rapamycin-conditioned CD4+ T cells can also act as next-generation cellular delivery vehicles-that is, "micropharmacies"-to disseminate corrective enzymes for multiple lysosomal storage disorders. We evaluated the therapeutic potential of this treatment platform for Fabry, Gaucher, Farber, and Pompe diseases in vitro and in vivo. For example, such micropharmacies expressing α-galactosidase A for treatment of Fabry disease were transplanted in mice where they provided functional enzyme in key affected tissues such as kidney and heart, facilitating clearance of pathogenic substrate after a single administration.
Collapse
Affiliation(s)
- Murtaza S Nagree
- Department of Medical BiophysicsUniversity of TorontoTorontoONCanada
- Department of PediatricsMedical College of WisconsinMilwaukeeWIUSA
| | | | - Mary L Faber
- Department of PediatricsMedical College of WisconsinMilwaukeeWIUSA
| | - Jitka Rybova
- Department of PediatricsMedical College of WisconsinMilwaukeeWIUSA
| | - C Anthony Rupar
- Department of Pathology and Laboratory MedicineWestern UniversityLondonONCanada
| | - S Ronan Foley
- Juravinski Hospital and Cancer CentreMcMaster UniversityHamiltonONCanada
| | - Maria Fuller
- Genetics and Molecular PathologySA Pathology at Women's and Children's HospitalNorth AdelaideSAAustralia
| | | | - Jeffrey A Medin
- Department of Medical BiophysicsUniversity of TorontoTorontoONCanada
- Department of PediatricsMedical College of WisconsinMilwaukeeWIUSA
- Department of BiochemistryMedical College of WisconsinMilwaukeeWIUSA
| |
Collapse
|
9
|
Abstract
PURPOSE OF REVIEW The aim of this study was to summarize recent findings in kidney gene therapy while proposing cystinuria as a model kidney disease target for genome engineering therapeutics. RECENT FINDINGS Despite the advances of gene therapy for treating diseases of other organs, the kidney lags behind. Kidney-targeted gene delivery remains an obstacle to gene therapy of kidney disease. Nanoparticle and adeno-associated viral vector technologies offer emerging hope for kidney gene therapy. Cystinuria represents a model potential target for kidney gene therapy due to its known genetic and molecular basis, targetability, and capacity for phenotypic rescue. SUMMARY Although gene therapy for kidney disease remains a major challenge, new and evolving technologies may actualize treatment for cystinuria and other kidney diseases.
Collapse
Affiliation(s)
- Jennifer L. Peek
- Medical Scientist Training Program, Vanderbilt University School of Medicine, Nashville, TN 37232
- Department of Pharmacology, Vanderbilt University, Nashville, TN 37232
| | - Matthew H. Wilson
- Department of Cell and Developmental Biology, Vanderbilt University, Nashville, TN 37232
- Department of Pharmacology, Vanderbilt University, Nashville, TN 37232
- Department of Medicine, Division of Nephrology and Hypertension, Vanderbilt University Medical Center, Nashville, TN 37232
- Department of Veterans Affairs, Tennessee Valley Health Services, Nashville, TN, 37212
| |
Collapse
|
10
|
Du Z, Ng YY, Zha S, Wang S. piggyBac system to co-express NKG2D CAR and IL-15 to augment the in vivo persistence and anti-AML activity of human peripheral blood NK cells. Mol Ther Methods Clin Dev 2021; 23:582-596. [PMID: 34853803 PMCID: PMC8609108 DOI: 10.1016/j.omtm.2021.10.014] [Citation(s) in RCA: 51] [Impact Index Per Article: 12.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2021] [Accepted: 10/29/2021] [Indexed: 12/26/2022]
Abstract
Promising progress has been made in adoptive transfer of allogeneic natural killer (NK) cells to treat relapsed or refractory acute myeloid leukemia (AML). In this regard, chimeric antigen receptor (CAR)-modification of NK cells is considered as a compelling approach to augment the specificity and cytotoxicity of NK cells against AML. Using a non-viral piggyBac transposon technology and human peripheral blood-derived primary NK cells, we generated CAR-NK cells to target NKG2D ligands and demonstrated their in vitro activity in lysing cancer cells expressing the ligands and in vivo efficacy in inhibiting tumor growth in a xenograft KG-1 AML model. We further generated CAR-NK cells co-expressing transgenes for the NKG2D CAR and interleukin-15 (IL-15). The ectopic expression of IL-15 improved the in vitro and in vivo persistence of NKG2D CAR-NK cells, leading to enhanced in vivo tumor control and significant prolongation of mouse survival in the KG-1 AML model. Collectively, our findings demonstrate the ectopic expression of IL-15 as an important means to improve the antileukemic activity of NKG2D CAR-NK cells. Our study further illustrates the feasibility of using the piggyBac non-viral platform as an efficient and cost-effective way for CAR-NK cell manufacturing.
Collapse
Affiliation(s)
- Zhicheng Du
- Department of Biological Sciences, National University of Singapore, 117543 Singapore, Singapore
| | - Yu Yang Ng
- Department of Biological Sciences, National University of Singapore, 117543 Singapore, Singapore
| | - Shijun Zha
- Department of Biological Sciences, National University of Singapore, 117543 Singapore, Singapore
| | - Shu Wang
- Department of Biological Sciences, National University of Singapore, 117543 Singapore, Singapore
| |
Collapse
|
11
|
Tay JC, Wang J, Du Z, Ng YY, Li Z, Ren Y, Zhang C, Zhu J, Xu XH, Wang S. Manufacturing NKG2D CAR-T cells with piggyBac transposon vectors and K562 artificial antigen-presenting cells. Mol Ther Methods Clin Dev 2021; 21:107-120. [PMID: 33816644 PMCID: PMC8005737 DOI: 10.1016/j.omtm.2021.02.023] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2020] [Accepted: 02/26/2021] [Indexed: 12/03/2022]
Abstract
Non-viral platforms can be applied rapidly and cost-effectively for chimeric antigen receptor (CAR)-T cell manufacturing. In the present paper, we describe in detail a clinically relevant manufacturing process for NKG2D CAR-T cells through electroporation of CAR-encoding piggyBac transposon plasmids and in vitro expansion with K562 artificial antigen-presenting cells. With an optimized protocol, we generated the final cell therapy products with 89.2% ± 10.2% NKG2D CAR-positive cells and achieved the corresponding antigen-dependent expansion between 50,000 and 60,000 folds within 4 weeks. To facilitate repeated CAR-T cell infusions, we evaluated the practicality of cryopreservation followed by post-thaw expansion and an extended manufacturing process for up to 9 rounds of weekly K562 cell stimulation. We found that neither compromised the in vitro anti-tumor activity of NKG2D CAR-T cells. Interestingly, the expression of T cell exhaustion markers TIGIT, TIM3, and LAG3 was reduced with extended manufacturing. To enhance the safety profile of the NKG2D CAR-T cells, we incorporated a full-length CD20 transgene in tandem with the CAR construct and demonstrated that autologous NK cells could mediate efficient antibody-dependent cell-mediated cytotoxicity to remove these CAR-T cells. Collectively, our study illustrates a protocol that generates large numbers of efficacious NKG2D CAR-T cells suitable for multiple rounds of infusions.
Collapse
Affiliation(s)
- Johan C.K. Tay
- Department of Biological Sciences, National University of Singapore, Singapore 117543, Singapore
| | - Junjian Wang
- Department of Gynaecologic Oncology, Cancer Hospital of the University of Chinese Academy of Sciences (Zhejiang Cancer Hospital), Hangzhou 310022, P.R. China
| | - Zhicheng Du
- Department of Biological Sciences, National University of Singapore, Singapore 117543, Singapore
| | - Yu Yang Ng
- Department of Biological Sciences, National University of Singapore, Singapore 117543, Singapore
| | - Zhendong Li
- Department of Biological Sciences, National University of Singapore, Singapore 117543, Singapore
| | - Yuefang Ren
- Department of Gynaecology, Huzhou Maternity & Child Health Care Hospital, Huzhou, Zhejiang 313000, P.R. China
| | - Chang Zhang
- Department of Gastrointestinal Surgery, The Third Affiliated Hospital of Guangzhou Medical University, Guangzhou 510150, P.R. China
| | - Jianqing Zhu
- Department of Gynaecologic Oncology, Cancer Hospital of the University of Chinese Academy of Sciences (Zhejiang Cancer Hospital), Hangzhou 310022, P.R. China
| | - Xue Hu Xu
- Department of Gastrointestinal Surgery, The Third Affiliated Hospital of Guangzhou Medical University, Guangzhou 510150, P.R. China
| | - Shu Wang
- Department of Biological Sciences, National University of Singapore, Singapore 117543, Singapore
| |
Collapse
|
12
|
Reinfeld BI, Madden MZ, Wolf MM, Chytil A, Bader JE, Patterson AR, Sugiura A, Cohen AS, Ali A, Do BT, Muir A, Lewis CA, Hongo RA, Young KL, Brown RE, Todd VM, Huffstater T, Abraham A, O'Neil RT, Wilson MH, Xin F, Tantawy MN, Merryman WD, Johnson RW, Williams CS, Mason EF, Mason FM, Beckermann KE, Vander Heiden MG, Manning HC, Rathmell JC, Rathmell WK. Cell-programmed nutrient partitioning in the tumour microenvironment. Nature 2021; 593:282-288. [PMID: 33828302 PMCID: PMC8122068 DOI: 10.1038/s41586-021-03442-1] [Citation(s) in RCA: 647] [Impact Index Per Article: 161.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2020] [Accepted: 03/10/2021] [Indexed: 02/01/2023]
Abstract
Cancer cells characteristically consume glucose through Warburg metabolism1, a process that forms the basis of tumour imaging by positron emission tomography (PET). Tumour-infiltrating immune cells also rely on glucose, and impaired immune cell metabolism in the tumour microenvironment (TME) contributes to immune evasion by tumour cells2-4. However, whether the metabolism of immune cells is dysregulated in the TME by cell-intrinsic programs or by competition with cancer cells for limited nutrients remains unclear. Here we used PET tracers to measure the access to and uptake of glucose and glutamine by specific cell subsets in the TME. Notably, myeloid cells had the greatest capacity to take up intratumoral glucose, followed by T cells and cancer cells, across a range of cancer models. By contrast, cancer cells showed the highest uptake of glutamine. This distinct nutrient partitioning was programmed in a cell-intrinsic manner through mTORC1 signalling and the expression of genes related to the metabolism of glucose and glutamine. Inhibiting glutamine uptake enhanced glucose uptake across tumour-resident cell types, showing that glutamine metabolism suppresses glucose uptake without glucose being a limiting factor in the TME. Thus, cell-intrinsic programs drive the preferential acquisition of glucose and glutamine by immune and cancer cells, respectively. Cell-selective partitioning of these nutrients could be exploited to develop therapies and imaging strategies to enhance or monitor the metabolic programs and activities of specific cell populations in the TME.
Collapse
Affiliation(s)
- Bradley I Reinfeld
- Medical Scientist Training Program, Vanderbilt University, Nashville, TN, USA
- Department of Medicine, Vanderbilt University Medical Center (VUMC), Nashville, TN, USA
- Graduate Program in Cancer Biology, Vanderbilt University, Nashville, TN, USA
| | - Matthew Z Madden
- Medical Scientist Training Program, Vanderbilt University, Nashville, TN, USA
- Department of Pathology, Microbiology and Immunology, VUMC, Nashville, TN, USA
| | - Melissa M Wolf
- Department of Medicine, Vanderbilt University Medical Center (VUMC), Nashville, TN, USA
- Graduate Program in Cancer Biology, Vanderbilt University, Nashville, TN, USA
| | - Anna Chytil
- Department of Medicine, Vanderbilt University Medical Center (VUMC), Nashville, TN, USA
| | - Jackie E Bader
- Department of Pathology, Microbiology and Immunology, VUMC, Nashville, TN, USA
| | - Andrew R Patterson
- Department of Pathology, Microbiology and Immunology, VUMC, Nashville, TN, USA
| | - Ayaka Sugiura
- Medical Scientist Training Program, Vanderbilt University, Nashville, TN, USA
- Department of Pathology, Microbiology and Immunology, VUMC, Nashville, TN, USA
| | - Allison S Cohen
- Department of Radiology and Radiological Sciences, VUMC, Nashville, TN, USA
- Vanderbilt University Institute of Imaging Science, VUMC, Nashville, TN, USA
| | - Ahmed Ali
- Koch Institute for Integrative Cancer Research and Department of Biology, Massachusetts Institute of Technology (MIT), Cambridge, MA, USA
- Broad Institute of MIT and Harvard University, Cambridge, MA, USA
| | - Brian T Do
- Koch Institute for Integrative Cancer Research and Department of Biology, Massachusetts Institute of Technology (MIT), Cambridge, MA, USA
- Broad Institute of MIT and Harvard University, Cambridge, MA, USA
| | - Alexander Muir
- Ben May Department for Cancer Research, University of Chicago, Chicago, IL, USA
| | - Caroline A Lewis
- Whitehead Institute for Biomedical Research, MIT, Cambridge, MA, USA
| | - Rachel A Hongo
- Department of Medicine, Vanderbilt University Medical Center (VUMC), Nashville, TN, USA
- Department of Pathology, Microbiology and Immunology, VUMC, Nashville, TN, USA
| | - Kirsten L Young
- Department of Medicine, Vanderbilt University Medical Center (VUMC), Nashville, TN, USA
- Department of Pathology, Microbiology and Immunology, VUMC, Nashville, TN, USA
| | - Rachel E Brown
- Medical Scientist Training Program, Vanderbilt University, Nashville, TN, USA
- Department of Medicine, Vanderbilt University Medical Center (VUMC), Nashville, TN, USA
- Graduate Program in Cancer Biology, Vanderbilt University, Nashville, TN, USA
| | - Vera M Todd
- Department of Medicine, Vanderbilt University Medical Center (VUMC), Nashville, TN, USA
- Graduate Program in Cancer Biology, Vanderbilt University, Nashville, TN, USA
| | - Tessa Huffstater
- Department of Biomedical Engineering, Vanderbilt University, Nashville, TN, USA
| | - Abin Abraham
- Medical Scientist Training Program, Vanderbilt University, Nashville, TN, USA
- Vanderbilt Genetics Institute, VUMC, Nashville, TN, USA
| | - Richard T O'Neil
- Department of Medicine, Vanderbilt University Medical Center (VUMC), Nashville, TN, USA
- Department of Veterans Affairs, Tennessee Valley Health System, Nashville, TN, USA
| | - Matthew H Wilson
- Department of Medicine, Vanderbilt University Medical Center (VUMC), Nashville, TN, USA
- Department of Veterans Affairs, Tennessee Valley Health System, Nashville, TN, USA
| | - Fuxue Xin
- Department of Radiology and Radiological Sciences, VUMC, Nashville, TN, USA
- Vanderbilt University Institute of Imaging Science, VUMC, Nashville, TN, USA
| | - M Noor Tantawy
- Department of Radiology and Radiological Sciences, VUMC, Nashville, TN, USA
- Vanderbilt University Institute of Imaging Science, VUMC, Nashville, TN, USA
| | - W David Merryman
- Department of Biomedical Engineering, Vanderbilt University, Nashville, TN, USA
| | - Rachelle W Johnson
- Department of Medicine, Vanderbilt University Medical Center (VUMC), Nashville, TN, USA
| | - Christopher S Williams
- Department of Medicine, Vanderbilt University Medical Center (VUMC), Nashville, TN, USA
- Department of Veterans Affairs, Tennessee Valley Health System, Nashville, TN, USA
| | - Emily F Mason
- Department of Pathology, Microbiology and Immunology, VUMC, Nashville, TN, USA
| | - Frank M Mason
- Department of Medicine, Vanderbilt University Medical Center (VUMC), Nashville, TN, USA
| | | | - Matthew G Vander Heiden
- Koch Institute for Integrative Cancer Research and Department of Biology, Massachusetts Institute of Technology (MIT), Cambridge, MA, USA
- Broad Institute of MIT and Harvard University, Cambridge, MA, USA
- Dana-Farber Cancer Institute, Boston, MA, USA
| | - H Charles Manning
- Department of Radiology and Radiological Sciences, VUMC, Nashville, TN, USA
- Vanderbilt University Institute of Imaging Science, VUMC, Nashville, TN, USA
| | - Jeffrey C Rathmell
- Department of Pathology, Microbiology and Immunology, VUMC, Nashville, TN, USA.
- Vanderbilt Center for Immunobiology and Vanderbilt-Ingram Cancer Center, VUMC, Nashville, TN, USA.
| | - W Kimryn Rathmell
- Department of Medicine, Vanderbilt University Medical Center (VUMC), Nashville, TN, USA.
- Vanderbilt Center for Immunobiology and Vanderbilt-Ingram Cancer Center, VUMC, Nashville, TN, USA.
| |
Collapse
|
13
|
Pozzi A, Harris RC. The Vanderbilt O'Brien Kidney Center. Am J Physiol Renal Physiol 2021; 320:F342-F350. [PMID: 33356958 PMCID: PMC7988814 DOI: 10.1152/ajprenal.00452.2020] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2020] [Revised: 12/14/2020] [Accepted: 12/14/2020] [Indexed: 11/22/2022] Open
Abstract
The Vanderbilt O'Brien Kidney Center (VOKC) is one of the eight National Institutes of Health P30-funded centers in the United States. The mission of these core-based centers is to provide technical and conceptual support to enhance and facilitate research in the field of kidney diseases. The goal of the VOKC is to provide support to understand mechanisms and identify potential therapies for acute and chronic kidney disease. The services provided by the VOKC are meant to help the scientific community to have the right support and tools as well as to select the right animal model, statistical analysis, and clinical study design to perform innovative research and translate discoveries into personalized care to prevent, diagnose, and cure kidney disease. To achieve these goals, the VOKC has in place a program to foster collaborative investigation into critical questions of kidney disease, to personalize diagnosis and treatment of kidney disease, and to disseminate information about kidney disease and the benefits of VOKC services and research. The VOKC is complemented by state-of-the-art cores and an education and outreach program whose goals are to provide an educational platform to enhance the study of kidney disease, to publicize information about services available through the VOKC, and to provide information about kidney disease to patients and other interested members of the community. In this review, we highlight the major services and contributions of the VOKC.
Collapse
Affiliation(s)
- Ambra Pozzi
- Division of Nephrology and Hypertension, Department of Medicine, Vanderbilt University School of Medicine, Nashville, Tennessee
- Vanderbilt Center for Kidney Disease, Vanderbilt University School of Medicine, Nashville, Tennessee
- Department of Veterans Affairs, Nashville, Tennessee
| | - Raymond C Harris
- Division of Nephrology and Hypertension, Department of Medicine, Vanderbilt University School of Medicine, Nashville, Tennessee
- Vanderbilt Center for Kidney Disease, Vanderbilt University School of Medicine, Nashville, Tennessee
- Department of Veterans Affairs, Nashville, Tennessee
| |
Collapse
|
14
|
Freen-van Heeren JJ. Using CRISPR to enhance T cell effector function for therapeutic applications. Cytokine X 2021; 3:100049. [PMID: 33604565 PMCID: PMC7885876 DOI: 10.1016/j.cytox.2020.100049] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2020] [Accepted: 12/15/2020] [Indexed: 12/12/2022] Open
Abstract
T cells are critical to fight pathogenic microbes and combat malignantly transformed cells in the fight against cancer. To exert their effector function, T cells produce effector molecules, such as the pro-inflammatory cytokines IFN-γ, TNF-α and IL-2. Tumors possess many inhibitory mechanisms that dampen T cell effector function, limiting the secretion of cytotoxic molecules. As a result, the control and elimination of tumors is impaired. Through recent advances in genomic editing, T cells can now be successfully modified via CRISPR/Cas9 technology. For instance, engaging (post-)transcriptional mechanisms to enhance T cell cytokine production, the retargeting of T cell antigen specificity or rendering T cells refractive to inhibitory receptor signaling can augment T cell effector function. Therefore, CRISPR/Cas9-mediated genome editing might provide novel strategies for cancer immunotherapy. Recently, the first-in-patient clinical trial was successfully performed with CRISPR/Cas9-modified human T cell therapy. In this review, a brief overview of currently available techniques is provided, and recent advances in T cell genomic engineering for the enhancement of T cell effector function for therapeutic purposes are discussed.
Collapse
Key Words
- AP-1, activator protein 1
- ARE, AU-rich element
- ARE-Del, deletion of the 3′UTR AREs from the Ifng/IFNG gene
- CAR T cells
- CAR, Chimeric Antigen Receptor
- CRISPR
- CRISPR, Clustered Regularly Interspaced Short Palindromic Repeat
- CRS, cytokine release syndrome
- CTLA-4, cytotoxic T-lymphocyte-associated protein 4
- Cas, CRISPR-associated
- Cas9
- Cytokines
- DGK, Diacylglycerol kinase
- DHX37, DEAH-box helicase 37
- EBV, Epstein Barr virus
- FOXP3, Forkhead box P3
- GATA, GATA binding protein
- Genome editing
- IFN, interferon
- IL, interleukin
- LAG-3, Lymphocyte Activating 3
- NF-κB, nuclear factor of activated B cells
- PD-1, Programmed cell Death 1
- PD-L1, Programmed Death Ligand 1
- PTPN2, Protein Tyrosine Phosphatase Non-Receptor 2
- Pdia3, Protein Disulfide Isomerase Family A Member 3
- RBP, RNA-binding protein
- RNP, ribonuclear protein
- T cell effector function
- T cells
- TCR, T cell receptor
- TGF, transforming growth factor
- TIL, Tumor Infiltrating Lymphocyte
- TLRs, Toll-like receptors
- TNF, tumor necrosis factor
- TRAC, TCR-α chain
- TRBC, TCR-β chain
- UTR, untranslated region
- tTCR, transgenic TCR
Collapse
|
15
|
Abdalla AME, Xiao L, Miao Y, Huang L, Fadlallah GM, Gauthier M, Ouyang C, Yang G. Nanotechnology Promotes Genetic and Functional Modifications of Therapeutic T Cells Against Cancer. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2020; 7:1903164. [PMID: 32440473 PMCID: PMC7237845 DOI: 10.1002/advs.201903164] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/14/2019] [Revised: 01/23/2020] [Indexed: 05/24/2023]
Abstract
Growing experience with engineered chimeric antigen receptor (CAR)-T cells has revealed some of the challenges associated with developing patient-specific therapy. The promising clinical results obtained with CAR-T therapy nevertheless demonstrate the urgency of advancements to promote and expand its uses. There is indeed a need to devise novel methods to generate potent CARs, and to confer them and track their anti-tumor efficacy in CAR-T therapy. A potentially effective approach to improve the efficacy of CAR-T cell therapy would be to exploit the benefits of nanotechnology. This report highlights the current limitations of CAR-T immunotherapy and pinpoints potential opportunities and tremendous advantages of using nanotechnology to 1) introduce CAR transgene cassettes into primary T cells, 2) stimulate T cell expansion and persistence, 3) improve T cell trafficking, 4) stimulate the intrinsic T cell activity, 5) reprogram the immunosuppressive cellular and vascular microenvironments, and 6) monitor the therapeutic efficacy of CAR-T cell therapy. Therefore, genetic and functional modifications promoted by nanotechnology enable the generation of robust CAR-T cell therapy and offer precision treatments against cancer.
Collapse
Affiliation(s)
- Ahmed M. E. Abdalla
- Department of Biomedical EngineeringCollege of Life Science and TechnologyHuazhong University of Science and TechnologyWuhan430074China
- Department of BiochemistryCollege of Applied ScienceUniversity of BahriKhartoum1660/11111Sudan
| | - Lin Xiao
- Department of Biomedical EngineeringCollege of Life Science and TechnologyHuazhong University of Science and TechnologyWuhan430074China
| | - Yu Miao
- Department of Vascular SurgeryGeneral Hospital of Ningxia Medical UniversityYinchuan750004China
| | - Lixia Huang
- Hubei Key Laboratory of Purification and Application of Plant Anti‐Cancer Active IngredientsSchool of Chemistry and Life SciencesHubei University of EducationWuhan430205China
| | - Gendeal M. Fadlallah
- Department of Chemistry and BiologyFaculty of EducationUniversity of GeziraWad‐Medani2667Sudan
| | - Mario Gauthier
- Department of ChemistryUniversity of WaterlooWaterlooN2L 3G1Canada
| | - Chenxi Ouyang
- Department of Vascular SurgeryFuwai HospitalNational Center for Cardiovascular DiseaseChinese Academy of Medical Sciences and Peking Union Medical CollegeBeijing100037China
| | - Guang Yang
- Department of Biomedical EngineeringCollege of Life Science and TechnologyHuazhong University of Science and TechnologyWuhan430074China
| |
Collapse
|
16
|
piggyBac-Based Non-Viral In Vivo Gene Delivery Useful for Production of Genetically Modified Animals and Organs. Pharmaceutics 2020; 12:pharmaceutics12030277. [PMID: 32204422 PMCID: PMC7151002 DOI: 10.3390/pharmaceutics12030277] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2020] [Revised: 03/17/2020] [Accepted: 03/18/2020] [Indexed: 11/17/2022] Open
Abstract
In vivo gene delivery involves direct injection of nucleic acids (NAs) into tissues, organs, or tail-veins. It has been recognized as a useful tool for evaluating the function of a gene of interest (GOI), creating models for human disease and basic research targeting gene therapy. Cargo frequently used for gene delivery are largely divided into viral and non-viral vectors. Viral vectors have strong infectious activity and do not require the use of instruments or reagents helpful for gene delivery but bear immunological and tumorigenic problems. In contrast, non-viral vectors strictly require instruments (i.e., electroporator) or reagents (i.e., liposomes) for enhanced uptake of NAs by cells and are often accompanied by weak transfection activity, with less immunological and tumorigenic problems. Chromosomal integration of GOI-bearing transgenes would be ideal for achieving long-term expression of GOI. piggyBac (PB), one of three transposons (PB, Sleeping Beauty (SB), and Tol2) found thus far, has been used for efficient transfection of GOI in various mammalian cells in vitro and in vivo. In this review, we outline recent achievements of PB-based production of genetically modified animals and organs and will provide some experimental concepts using this system.
Collapse
|
17
|
Shiina T, Blancher A. The Cynomolgus Macaque MHC Polymorphism in Experimental Medicine. Cells 2019; 8:E978. [PMID: 31455025 PMCID: PMC6770713 DOI: 10.3390/cells8090978] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2019] [Revised: 08/20/2019] [Accepted: 08/22/2019] [Indexed: 02/07/2023] Open
Abstract
Among the non-human primates used in experimental medicine, cynomolgus macaques (Macaca fascicularis hereafter referred to as Mafa) are increasingly selected for the ease with which they are maintained and bred in captivity. Macaques belong to Old World monkeys and are phylogenetically much closer to humans than rodents, which are still the most frequently used animal model. Our understanding of the Mafa genome has progressed rapidly in recent years and has greatly benefited from the latest technical advances in molecular genetics. Cynomolgus macaques are widespread in Southeast Asia and numerous studies have shown a distinct genetic differentiation of continental and island populations. The major histocompatibility complex of cynomolgus macaque (Mafa MHC) is organized in the same way as that of human, but it differs from the latter by its high degree of classical class I gene duplication. Human polymorphic MHC regions play a pivotal role in allograft transplantation and have been associated with more than 100 diseases and/or phenotypes. The Mafa MHC polymorphism similarly plays a crucial role in experimental allografts of organs and stem cells. Experimental results show that the Mafa MHC class I and II regions influence the ability to mount an immune response against infectious pathogens and vaccines. MHC also affects cynomolgus macaque reproduction and impacts on numerous biological parameters. This review describes the Mafa MHC polymorphism and the methods currently used to characterize it. We discuss some of the major areas of experimental medicine where an effect induced by MHC polymorphism has been demonstrated.
Collapse
Affiliation(s)
- Takashi Shiina
- Department of Molecular Life Sciences, Division of Basic Medical Science and Molecular Medicine, Tokai University School of Medicine, 143 Shimokasuya, Isehara, Kanagawa 259-1193, Japan
| | - Antoine Blancher
- Centre de Physiopathologie Toulouse-Purpan (CPTP), Université de Toulouse, Centre National de la Recherche Scientifique (CNRS), Institut National de la Santé et de la Recherche Médicale (Inserm), Université Paul Sabatier (UPS), Toulouse 31000, France.
- Laboratoire d'immunologie, CHU de Toulouse, Institut Fédératif de Biologie, hôpital Purpan, 330 Avenue de Grande Bretagne, TSA40031, 31059 Toulouse CEDEX 9, France.
| |
Collapse
|
18
|
Ghezlou M, Mokhtari F, Kalbasi A, Riazi G, Kaghazian H, Emadi R, Aref AR. Aggregate Forms of Recombinant Human Erythropoietin With Different Charge Profile Substantially Impact Biological Activities. J Pharm Sci 2019; 109:277-283. [PMID: 31216452 DOI: 10.1016/j.xphs.2019.05.036] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2019] [Revised: 05/13/2019] [Accepted: 05/24/2019] [Indexed: 11/17/2022]
Abstract
Recombinant human erythropoietin (rHuEPO) as a glycoprotein growth factor has been considered a biological drug for treatment of anemic patients with chronic renal failure or who receive cancer chemotherapy. Biological activity and circulation time are 2 parameters that are important to achieve EPO's efficacy. Previous efforts for increasing EPO's efficacy have focused on glycosylation modification via adding more sialic acid antenna and generates more negative charged protein. Evidences cleared that EPO's activity increased by numbers of N-glycan moieties with presence of sialic acids at their terminus. Correlation between bioactivity and glycosylation with terminal sialylation is theoretically achieved using the calculation of the amount of charge profile of the EPO variants called "I-number." Here, we studied and compared the relationship between bioactivities of different EPOs that contained various I-numbers and the effect of their secondary and tertiary protein structures on measured in vivo efficacy. Eight recombinant EPOs batches were produced under the same condition. I-numbers found out by EPO's charge profiles determination using capillary electrophoresis and activities were studied upon erythroid precursor cell stimulation in mice. Analyzing the bioactivity, I-number, and structural studies revealed that in spite of I-number, conformational changes in protein structure and presence of aggregated species impact bioactivity substantially.
Collapse
Affiliation(s)
- Mansoureh Ghezlou
- Biophysics Department, Science and Research Branch, Islamic Azad University, Tehran, Iran; Bioorganic Lab, Biochemistry Department, University of Tehran, Tehran, Iran
| | - Farzad Mokhtari
- Bioorganic Lab, Biochemistry Department, University of Tehran, Tehran, Iran
| | - Alireza Kalbasi
- Department of Surgery, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, Massachusetts 02215.
| | | | - Hooman Kaghazian
- Department of Recombinant Biopharmaceutical Production, Research and Production Complex, Pasteur Institute of Iran, Tehran, Iran
| | - Reza Emadi
- Bioorganic Lab, Biochemistry Department, University of Tehran, Tehran, Iran
| | - Amir Reza Aref
- Belfer Center for Applied Cancer Science, Dana-Farber Cancer Institute, Harvard Medical School, Boston, Massachusetts 02215.
| |
Collapse
|
19
|
Hajimohammadjafartehrani M, Hosseinali SH, Dehkohneh A, Ghoraeian P, Ale-Ebrahim M, Akhtari K, Shahpasand K, Saboury AA, Attar F, Falahati M. The effects of nickel oxide nanoparticles on tau protein and neuron-like cells: Biothermodynamics and molecular studies. Int J Biol Macromol 2019; 127:330-339. [DOI: 10.1016/j.ijbiomac.2019.01.050] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2018] [Revised: 12/26/2018] [Accepted: 01/11/2019] [Indexed: 12/13/2022]
|