1
|
Kushwaha N, Panjwani D, Patel S, Ahlawat P, Yadav MR, Patel AS. Emerging advances in nano-biomaterial assisted amyloid beta chimeric antigen receptor macrophages (CAR-M) therapy: reducing plaque burden in Alzheimer's disease. J Drug Target 2025; 33:185-205. [PMID: 39403775 DOI: 10.1080/1061186x.2024.2417012] [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: 08/04/2024] [Revised: 09/23/2024] [Accepted: 10/08/2024] [Indexed: 10/22/2024]
Abstract
Alzheimer's disease is the most common form, accounting for 60-70% of 55 million dementia cases. Even though the precise pathophysiology of AD is not completely understood, clinical trials focused on antibodies targeting aggregated forms of β amyloid (Aβ) have demonstrated that reducing amyloid plaques can arrest cognitive decline in patients in the early stages of AD. In this study, we provide an overview of current research and innovations for controlled release from nano-biomaterial-assisted chimeric antigen receptor macrophage (CAR-M) therapeutic strategies targeted at AD. Nano-bio materials, such as iron-oxide nanoparticles (IONPs), can be made selectively (Hp-Hb/mannose) to bind and take up Aβ plaques like CAR-M cells. By using nano-bio materials, both the delivery and stability of CAR-M cells in brain tissue can be improved to overcome the barriers of the BBB and enhance therapeutic effects. By enhancing the targeting capabilities and stability of CAR-M cells, mRNA-loaded nano-biomaterials can significantly improve the efficacy of immunotherapy for plaque reduction in AD. This novel strategy holds promise for translating preclinical successes into clinical applications, potentially revolutionising the management of AD.
Collapse
Affiliation(s)
- Nishabh Kushwaha
- Department of Pharmaceutics, Parul Institute of Pharmacy, Parul University, Vadodara, India
| | - Drishti Panjwani
- Department of Pharmaceutics, Parul Institute of Pharmacy, Parul University, Vadodara, India
| | - Shruti Patel
- Department of Pharmaceutics, Parul Institute of Pharmacy, Parul University, Vadodara, India
| | - Priyanka Ahlawat
- Department of Pharmaceutics, Parul Institute of Pharmacy, Parul University, Vadodara, India
| | - Mange Ram Yadav
- Research and Development Cell, Parul University, Vadodara, India
| | - Asha S Patel
- Department of Pharmaceutics, Parul Institute of Pharmacy, Parul University, Vadodara, India
| |
Collapse
|
2
|
Ahmed HM, Moselhy SS, Mohamad MI, Soliman AF, Hassan MNM, El-Khazragy N. Targeting refractory diffuse large B cell lymphoma by CAR-WEE1 T-cells: In vitro evaluation. Ann Hematol 2025:10.1007/s00277-024-06134-8. [PMID: 39820427 DOI: 10.1007/s00277-024-06134-8] [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: 07/06/2024] [Accepted: 12/02/2024] [Indexed: 01/19/2025]
Abstract
Refractory Diffuse Large B-cell Lymphoma (DLBCL) presents a major therapeutic challenge due to its resistance to standard treatments. Engineered T-cells, especially Chimeric Antigen Receptor (CAR) T-cells, have shown promise in overcoming drug resistance. This study investigates the effectiveness of WEE1-engineered T-cells in targeting and eliminating refractory DLBCL in vitro. CAR T-cells were created by transducing a 5th-generation CAR construct designed to recognize WEE1, a surface antigen commonly found on refractory DLBCL cells. The cytotoxic effect of engineered T-cells was tested against Rituximab-resistant DLBCL cells (RR-NU-DUL-1). Apoptosis and cell cycle were evaluated using flow cytometry. Quantitative Real-time PCR (RT-PCR) was used to measure the expression of WEE1, BCL2, and CDK2. The results showed a significant increase in target cell lysis, apoptosis, and necrosis, a significant reduction in the percentage of cells in the G2M phase of the cell cycle, as well as a decrease in gene expression level, indicating strong anti-tumor activity. These findings suggest that CAR T-cell therapy holds great promise for treating refractory DLBCL, offering a potential path for clinical application. This in vitro evaluation highlights the potential of WEE1-engineered T-cells as a targeted treatment strategy for refractory DLBCL, emphasizing their clinical applicability and ability to overcome resistance mechanisms in this aggressive lymphoma subtype.
Collapse
Affiliation(s)
- Hadeer Mohamed Ahmed
- Department of Biochemistry, Faculty of Science, Ain Shams University, Cairo, 11566, Egypt
| | - Said Salama Moselhy
- Department of Biochemistry, Faculty of Science, Ain Shams University, Cairo, 11566, Egypt
| | - Magda I Mohamad
- Department of Medical Biochemistry and Molecular Biology, Faculty of Medicine, Ain Shams University, Cairo, 11566, Egypt
| | - Ahmed F Soliman
- Department of Biochemistry, Faculty of Science, Ain Shams University, Cairo, 11566, Egypt
| | - Marwa N M Hassan
- Department of Medical Biochemistry and Molecular Biology, Faculty of Medicine, Ain Shams University, Cairo, 11566, Egypt
| | - Nashwa El-Khazragy
- Department of Clinical Pathology-Hematology and AinShams Medical Research Institute (MASRI), Faculty of Medicine, Ain Shams University, Cairo, 11566, Egypt.
| |
Collapse
|
3
|
Lam PY, Omer N, Wong JKM, Tu C, Alim L, Rossi GR, Victorova M, Tompkins H, Lin C, Mehdi AM, Choo A, Elliott MR, Coleborn E, Sun J, Mercer T, Vittorio O, Dobson LJ, McLellan AD, Brooks A, Tuong ZK, Cheetham SW, Nicholls W, Souza‐Fonseca‐Guimaraes F. Enhancement of anti-sarcoma immunity by NK cells engineered with mRNA for expression of a EphA2-targeted CAR. Clin Transl Med 2025; 15:e70140. [PMID: 39763064 PMCID: PMC11705447 DOI: 10.1002/ctm2.70140] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2024] [Revised: 11/30/2024] [Accepted: 12/06/2024] [Indexed: 01/11/2025] Open
Abstract
BACKGROUND Paediatric sarcomas, including rhabdomyosarcoma, Ewing sarcoma and osteosarcoma, represent a group of malignancies that significantly contribute to cancer-related morbidity and mortality in children and young adults. These cancers share common challenges, including high rates of metastasis, recurrence or treatment resistance, leading to a 5-year survival rate of approximately 20% for patients with advanced disease stages. Despite the critical need, therapeutic advancements have been limited over the past three decades. The advent of chimeric antigen receptor (CAR)-based immunotherapies offers a promising avenue for novel treatments. However, CAR-T cells have faced significant challenges and limited success in treating solid tumours due to issues such as poor tumour infiltration, immunosuppressive tumour microenvironments and off-target effects. In contrast, the adaptation of CAR technology for natural killer (NK) cells has demonstrated potential in both haematological and solid tumours, suggesting a new therapeutic strategy for paediatric sarcomas. METHODS This study developed and validated a novel CAR-NK cell therapy targeting the ephrin type-A receptor-2 (EphA2) antigen, which is highly expressed in various paediatric sarcomas. RESULTS CAR expression was successfully detected on the surface of NK cells post-electroporation, indicating successful transfection. Significantly, EphA2-specific CAR-NK cells demonstrated enhanced cytotoxic activity against several paediatric sarcoma cell lines in vitro, including those of rhabdomyosarcoma, Ewing sarcoma and osteosarcoma, compared to unmodified NK cells. Transient messenger RNA (mRNA) transfection of NK cells is a safe approach in genetic engineering, with further chemical modifications to mRNA enhancing stability of temporal EphA2-CAR expression in NK cells, thereby promoting prolonged protein expression. Additionally, in vivo EphA2-CAR-NK cells showed promising anti-cancer activity in rhabdomyosarcoma and osteosarcoma mouse models. CONCLUSIONS The study provides a foundational basis for the clinical evaluation of EphA2-targeted CAR-NK cell therapy across a spectrum of paediatric sarcomas. The enhanced anti-tumour effects observed in vitro/vivo suggests potential for improved therapeutic outcomes in hard-to-cure paediatric sarcomas. KEY POINTS Addressing unmet clinical needs in paediatric Sarcomas. Paediatric sarcomas, including rhabdomyosarcoma, Ewing sarcoma, and osteosarcoma, exhibit poor survival rates in advanced disease stages. The lack of significant therapeutic progress over the past three decades necessitates innovative treatment approaches. Advancing immunotherapy with CAR-NK cells. Natural killer (NK) cells modified with chimeric antigen receptors (CARs) represent a promising strategy to overcome the limitations of CAR-T cells, particularly in solid tumours. CAR-NK cells are associated with enhanced tumour targeting, reduced off-target effects, and improved safety profiles. EphA2 as a therapeutic target. EphA2, a receptor overexpressed in multiple paediatric sarcomas, is identified as a viable target for CAR-based immunotherapy due to its critical role in tumour progression and angiogenesis. Innovations in mRNA-based engineering. This study demonstrates the feasibility of transient mRNA transfection to engineer NK cells for CAR expression, offering a non-integrative and safer alternative to viral transduction. Enhancements in mRNA stability through chemical modifications, can further optimise protein expression. Preclinical efficacy of EphA2-CAR NK cells. EphA2-specific CAR-NK cells exhibit superior cytotoxicity against sarcoma cell lines in vitro and demonstrate significant anti-tumour activity in in vivo mouse models of rhabdomyosarcoma and osteosarcoma. Clinical translation potential. The findings establish a strong preclinical rationale for the clinical evaluation of EphA2-targeted CAR-NK therapy as a novel immunotherapeutic option for paediatric sarcomas. Future research directions: Combining EphA2-CAR NK cells with immune checkpoint inhibitors or other immunomodulatory agents could further enhance therapeutic outcomes and durability. Advanced preclinical models mimicking human tumour microenvironments are needed to refine and optimise this therapeutic approach.
Collapse
MESH Headings
- Killer Cells, Natural/immunology
- Killer Cells, Natural/metabolism
- Receptor, EphA2/genetics
- Humans
- Animals
- Sarcoma/therapy
- Sarcoma/immunology
- Sarcoma/genetics
- Mice
- Receptors, Chimeric Antigen/genetics
- Receptors, Chimeric Antigen/immunology
- RNA, Messenger/genetics
- RNA, Messenger/metabolism
- Immunotherapy, Adoptive/methods
- Cell Line, Tumor
- Disease Models, Animal
Collapse
Affiliation(s)
- Pui Yeng Lam
- Frazer Institute, Faculty of MedicineThe University of QueenslandWoolloongabbaQueenslandAustralia
| | - Natacha Omer
- Frazer Institute, Faculty of MedicineThe University of QueenslandWoolloongabbaQueenslandAustralia
- Queensland Children's HospitalBrisbaneQueenslandAustralia
| | - Josh K. M. Wong
- Frazer Institute, Faculty of MedicineThe University of QueenslandWoolloongabbaQueenslandAustralia
| | - Cui Tu
- Frazer Institute, Faculty of MedicineThe University of QueenslandWoolloongabbaQueenslandAustralia
| | - Louisa Alim
- Frazer Institute, Faculty of MedicineThe University of QueenslandWoolloongabbaQueenslandAustralia
| | - Gustavo R. Rossi
- Frazer Institute, Faculty of MedicineThe University of QueenslandWoolloongabbaQueenslandAustralia
| | - Maria Victorova
- Australian Institute for Bioengineering and NanotechnologyUniversity of QueenslandSt LuciaQueenslandAustralia
- BASE FacilityUniversity of QueenslandSt LuciaQueenslandAustralia
| | - Hannah Tompkins
- BASE FacilityUniversity of QueenslandSt LuciaQueenslandAustralia
| | - Cheng‐Yu Lin
- Frazer Institute, Faculty of MedicineThe University of QueenslandWoolloongabbaQueenslandAustralia
| | - Ahmed M. Mehdi
- Frazer Institute, Faculty of MedicineThe University of QueenslandWoolloongabbaQueenslandAustralia
- Queensland Cyber Infrastructure Foundation Ltd (QCIF)Facility for Advanced BioinformaticsSt LuciaQueenslandAustralia
| | - Amos Choo
- Ian Frazer Centre for Children's Immunotherapy Research, Child Health Research Centre, Faculty of MedicineThe University of QueenslandBrisbaneQueenslandAustralia
| | - Melissa R. Elliott
- Frazer Institute, Faculty of MedicineThe University of QueenslandWoolloongabbaQueenslandAustralia
| | - Elaina Coleborn
- Frazer Institute, Faculty of MedicineThe University of QueenslandWoolloongabbaQueenslandAustralia
| | - Jane Sun
- Frazer Institute, Faculty of MedicineThe University of QueenslandWoolloongabbaQueenslandAustralia
| | - Timothy Mercer
- Australian Institute for Bioengineering and NanotechnologyUniversity of QueenslandSt LuciaQueenslandAustralia
- BASE FacilityUniversity of QueenslandSt LuciaQueenslandAustralia
| | - Orazio Vittorio
- School of Biomedical Sciences, Faculty of Medicine and HealthUniversity of New South WalesSydneyNew South WalesAustralia
| | - Lachlan J. Dobson
- Department of Microbiology and ImmunologyThe University of OtagoDunedinNew Zealand
| | | | - Andrew Brooks
- Frazer Institute, Faculty of MedicineThe University of QueenslandWoolloongabbaQueenslandAustralia
| | - Zewen Kelvin Tuong
- Frazer Institute, Faculty of MedicineThe University of QueenslandWoolloongabbaQueenslandAustralia
- Ian Frazer Centre for Children's Immunotherapy Research, Child Health Research Centre, Faculty of MedicineThe University of QueenslandBrisbaneQueenslandAustralia
| | - Seth W. Cheetham
- Australian Institute for Bioengineering and NanotechnologyUniversity of QueenslandSt LuciaQueenslandAustralia
- BASE FacilityUniversity of QueenslandSt LuciaQueenslandAustralia
| | - Wayne Nicholls
- Queensland Children's HospitalBrisbaneQueenslandAustralia
- Ian Frazer Centre for Children's Immunotherapy Research, Child Health Research Centre, Faculty of MedicineThe University of QueenslandBrisbaneQueenslandAustralia
| | | |
Collapse
|
4
|
Padzińska-Pruszyńska IB, Taciak B, Kiraga Ł, Smolarska A, Górczak M, Kucharzewska P, Kubiak M, Szeliga J, Matejuk A, Król M. Targeting Cancer: Microenvironment and Immunotherapy Innovations. Int J Mol Sci 2024; 25:13569. [PMID: 39769334 PMCID: PMC11679359 DOI: 10.3390/ijms252413569] [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: 11/13/2024] [Revised: 12/12/2024] [Accepted: 12/16/2024] [Indexed: 01/11/2025] Open
Abstract
In 2024, the United States was projected to experience 2 million new cancer diagnoses and approximately 611,720 cancer-related deaths, reflecting a broader global trend in which cancer cases are anticipated to exceed 35 million by 2050. This increasing burden highlights ongoing challenges in cancer treatment despite significant advances that have reduced cancer mortality by 31% since 1991. Key obstacles include the disease's inherent heterogeneity and complexity, such as treatment resistance, cancer stem cells, and the multifaceted tumor microenvironment (TME). The TME-comprising various tumor and immune cells, blood vessels, and biochemical factors-plays a crucial role in tumor growth and resistance to therapies. Recent innovations in cancer treatment, particularly in the field of immuno-oncology, have leveraged insights into TME interactions. An emerging example is the FDA-approved therapy using tumor-infiltrating lymphocytes (TILs), demonstrating the potential of cell-based approaches in solid tumors. However, TIL therapy is just one of many strategies being explored. This review provides a comprehensive overview of the emerging field of immuno-oncology, focusing on how novel therapies targeting or harnessing components of the TME could enhance treatment efficacy and address persistent challenges in cancer care.
Collapse
Affiliation(s)
- Irena Barbara Padzińska-Pruszyńska
- Center of Cellular Immunotherapies, Warsaw University of Life Sciences, 02-787 Warsaw, Poland; (I.B.P.-P.); (B.T.); (A.S.); (M.G.); (P.K.); (M.K.); (J.S.)
| | - Bartłomiej Taciak
- Center of Cellular Immunotherapies, Warsaw University of Life Sciences, 02-787 Warsaw, Poland; (I.B.P.-P.); (B.T.); (A.S.); (M.G.); (P.K.); (M.K.); (J.S.)
| | - Łukasz Kiraga
- Division of Pharmacology and Toxicology, Department of Preclinical Sciences, Institute of Veterinary Medicine, Warsaw University of Life Sciences, 02-787 Warsaw, Poland;
| | - Anna Smolarska
- Center of Cellular Immunotherapies, Warsaw University of Life Sciences, 02-787 Warsaw, Poland; (I.B.P.-P.); (B.T.); (A.S.); (M.G.); (P.K.); (M.K.); (J.S.)
| | - Małgorzata Górczak
- Center of Cellular Immunotherapies, Warsaw University of Life Sciences, 02-787 Warsaw, Poland; (I.B.P.-P.); (B.T.); (A.S.); (M.G.); (P.K.); (M.K.); (J.S.)
| | - Paulina Kucharzewska
- Center of Cellular Immunotherapies, Warsaw University of Life Sciences, 02-787 Warsaw, Poland; (I.B.P.-P.); (B.T.); (A.S.); (M.G.); (P.K.); (M.K.); (J.S.)
| | - Małgorzata Kubiak
- Center of Cellular Immunotherapies, Warsaw University of Life Sciences, 02-787 Warsaw, Poland; (I.B.P.-P.); (B.T.); (A.S.); (M.G.); (P.K.); (M.K.); (J.S.)
| | - Jacek Szeliga
- Center of Cellular Immunotherapies, Warsaw University of Life Sciences, 02-787 Warsaw, Poland; (I.B.P.-P.); (B.T.); (A.S.); (M.G.); (P.K.); (M.K.); (J.S.)
| | - Agata Matejuk
- Department of Immunology, Collegium Medicum, University of Zielona Góra, 65-046 Zielona Góra, Poland;
| | - Magdalena Król
- Center of Cellular Immunotherapies, Warsaw University of Life Sciences, 02-787 Warsaw, Poland; (I.B.P.-P.); (B.T.); (A.S.); (M.G.); (P.K.); (M.K.); (J.S.)
| |
Collapse
|
5
|
Alsuliman T, Marjanovic Z, Rimar D, Tarte K, Avcin T, Hagen M, Schett G, Farge D. Harnessing the potential of CAR-T cell in lupus treatment: From theory to practice. Autoimmun Rev 2024; 23:103687. [PMID: 39532175 DOI: 10.1016/j.autrev.2024.103687] [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: 08/28/2024] [Revised: 10/28/2024] [Accepted: 11/08/2024] [Indexed: 11/16/2024]
Abstract
Systemic Lupus Erythematosus (SLE) is a rare, heterogeneous, potentially life-threatening autoimmune disease. Presence of kidney or other major organ (brain, heart or lung) involvement are predictors of poor outcome and in a subset of patients resistant to 1st or 2nd line conventional treatment. The 10-year mortality remains around 10-15 %. Chimeric Antigen Receptors (CAR) are molecules that allow to redirect the engineered immune cells towards specific target antigens and to simultaneously boost their activation. Following breakthrough results observed in the treatment of hematological malignancies, conventional CAR T-cell therapy has recently been applied to refractory SLE patients. Compared to the use of monoclonal antibodies, anti-CD19 CAR T-cells allow to achieve deeper depletion of autoreactive B cells, notably at site of inflamed tissues and lymphoid organs (i.e. lymph node), to suppress interferon signature and to restore the immune tolerance with the reemergence of naïve B-cells with a new repertoire. All clinical data reported in SLE patients so far showed that autologous anti-CD19 CAR T-cell treatment allowed impressive short- and longer-term resolution of lupus nephritis and other severe disease-related manifestations, without major toxicities and only mild cytokine-release syndrome. These clinical effects persisted after B-cell reconstitution and were associated with normalization of double-stranded DNA antibodies and complement levels in drug-free patients until three years after the procedure. Overall, these pioneering experiences show unique clinical and immunological response to CAR T-cell therapy in SLE, and the need for extended follow-up to determine its long-term efficacy.
Collapse
Affiliation(s)
- Tamim Alsuliman
- Saint-Antoine Hospital, AP-HP, Service d'hématologie et thérapie cellulaire, 75012 Paris, France; Sorbonne University, Paris, France
| | - Zora Marjanovic
- Saint-Antoine Hospital, AP-HP, Service d'hématologie et thérapie cellulaire, 75012 Paris, France; Sorbonne University, Paris, France
| | - Doron Rimar
- Rheumatology unit, Bnai Zion medical center, Haifa, Israel; Ruth and Bruce Rappaport Faculty of Medicine, Technion, Haifa, Israel
| | - Karin Tarte
- SITI, CHU Rennes, UMR 1236, Université Rennes, INSERM, Etablissement Français du Sang Bretagne, F-35000 Rennes, France
| | - Tadej Avcin
- Department of Allergology, Rheumatology and Clinical Immunology, University Children's Hospital, University Medical Center Ljubljana and Department of Pediatrics, Faculty of Medicine, University of Ljubljana, Slovenia
| | - Melanie Hagen
- Department of Internal Medicine 3 - Rheumatology and Immunology, FAU Erlangen-Nürnberg and Universitätsklinikum Erlangen, Erlangen, Germany
| | - Georg Schett
- Department of Internal Medicine 3 - Rheumatology and Immunology, FAU Erlangen-Nürnberg and Universitätsklinikum Erlangen, Erlangen, Germany
| | - Dominique Farge
- Internal Medicine Unit (04): CRMR MATHEC, Maladies Auto-immunes et Thérapie Cellulaire, Centre de Référence des Maladies auto-immunes systémiques Rares d'Ile-de-France, AP-HP, St-Louis Hospital, France; Paris-Cité University, IRSL, Recherche clinique appliquée à l'hématologie, URP 3518, F-75010 Paris, France; Department of Medicine, McGill University, Montreal, QC, Canada.
| |
Collapse
|
6
|
Mirvis E, Benjamin R. Are we there yet? CAR-T therapy in multiple myeloma. Br J Haematol 2024; 205:2175-2189. [PMID: 39558776 PMCID: PMC11637742 DOI: 10.1111/bjh.19896] [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: 08/15/2024] [Accepted: 10/31/2024] [Indexed: 11/20/2024]
Abstract
The last few years have seen a revolution in cellular immunotherapies for multiple myeloma (MM) with novel antigen targets. The principle new target is B-cell maturation antigen (BCMA). Autologous chimeric antigen receptor T-cell (CAR-T) therapy directed against BCMA was first approved by the US Food and Drug Administration (FDA) and European Medicines Agency (EMA) in 2021, although approval by the National Institute for Health and Care Excellent (NICE) is awaited. Initial response rates in patients with heavily pretreated MM have been impressive, but patients are still relapsing. Furthermore, CAR-T manufacturing is expensive and time-consuming, and T-cell fitness is impaired by prior MM treatment. Numerous strategies to improve outcomes and delivery of cellular immunotherapy are under investigation, including next-generation CARs, allogeneic 'off-the-shelf' CARs and targeting of other MM antigens including G protein-coupled receptor, class C, group 5, member D (GPRC5D), Fc receptor homologue 5 (FcRH5), cluster of differentiation (CD)19, signalling lymphocyte activation molecule family member 7 (SLAMF7) and several others. In this exciting and rapidly evolving treatment landscape, this review evaluates the most recent clinical and preclinical data pertaining to these new cellular immunotherapies and explores strategies to overcome resistance pathways. On the protracted journey to a long-term cure, we outline the challenges that lie ahead and ask, 'Are we there yet?'
Collapse
Affiliation(s)
- Eitan Mirvis
- School of Cancer & Pharmaceutical Sciences, King's College LondonLondonUK
- Department of HaematologyKing's College Hospital NHS Foundation TrustLondonUK
| | - Reuben Benjamin
- School of Cancer & Pharmaceutical Sciences, King's College LondonLondonUK
- Department of HaematologyKing's College Hospital NHS Foundation TrustLondonUK
| |
Collapse
|
7
|
Carcopino C, Erdogan E, Henrich M, Kobold S. Armoring chimeric antigen receptor (CAR) T cells as micropharmacies for cancer therapy. IMMUNO-ONCOLOGY TECHNOLOGY 2024; 24:100739. [PMID: 39711794 PMCID: PMC11659983 DOI: 10.1016/j.iotech.2024.100739] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/24/2024]
Abstract
Chimeric antigen receptor (CAR)-T-cell therapy has emerged as a powerful weapon in the fight against cancer. However, its efficacy is often hindered by challenges such as limited tumor penetration, antigen escape, and immune suppression within the tumor microenvironment. This review explores the potential of armored CAR-T cells, or 'micropharmacies', in overcoming these obstacles and enhancing the therapeutic outcomes of adoptive T-cell (ATC) therapy. We delve into the engineering strategies behind these advanced therapies and the mechanisms through which they improve CAR-T-cell efficacy. Additionally, we discuss the latest advancements and research findings in the field, providing a comprehensive understanding of the role of armored CAR-T cells in cancer treatment. Ultimately, this review highlights the promising future of integrating micropharmacies into ATC therapy, paving the way for more effective and targeted cancer treatments.
Collapse
Affiliation(s)
- C. Carcopino
- Division of Clinical Pharmacology, Department of Medicine IV, University Hospital, Ludwig Maximilian University (LMU) of Munich, Munich, Germany
| | - E. Erdogan
- Division of Clinical Pharmacology, Department of Medicine IV, University Hospital, Ludwig Maximilian University (LMU) of Munich, Munich, Germany
| | - M. Henrich
- Division of Clinical Pharmacology, Department of Medicine IV, University Hospital, Ludwig Maximilian University (LMU) of Munich, Munich, Germany
| | - S. Kobold
- Division of Clinical Pharmacology, Department of Medicine IV, University Hospital, Ludwig Maximilian University (LMU) of Munich, Munich, Germany
- German Cancer Consortium (DKTK), Partner Site Munich, a partnership between the DKFZ Heidelberg and the University Hospital of the LMU, Heidelberg, Germany
- Einheit für Klinische Pharmakologie (EKLiP), Helmholtz Zentrum München - German Research Center for Environmental Health, Neuherberg, Germany
| |
Collapse
|
8
|
Nolan-Stevaux O, Smith R. Logic-gated and contextual control of immunotherapy for solid tumors: contrasting multi-specific T cell engagers and CAR-T cell therapies. Front Immunol 2024; 15:1490911. [PMID: 39606234 PMCID: PMC11599190 DOI: 10.3389/fimmu.2024.1490911] [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: 09/03/2024] [Accepted: 10/18/2024] [Indexed: 11/29/2024] Open
Abstract
CAR-T cell and T cell engager therapies have demonstrated transformational efficacy against hematological malignancies, but achieving efficacy in solid tumors has been more challenging, in large part because of on-target/off-tumor toxicities and sub-optimal T cell anti-tumor cytotoxic functions. Here, we discuss engineering solutions that exploit biological properties of solid tumors to overcome these challenges. Using logic gates as a framework, we categorize the numerous approaches that leverage two inputs instead of one to achieve better cancer selectivity or efficacy in solid tumors with dual-input CAR-Ts or multi-specific TCEs. In addition to the "OR gate" and "AND gate" approaches that leverage dual tumor antigen targeting, we also review "contextual AND gate" technologies whereby continuous cancer-selective inputs such a pH, hypoxia, target density, tumor proteases, and immune-suppressive cytokine gradients can be creatively incorporated in therapy designs. We also introduce the notion of "output directionality" to distinguish dual-input strategies that mechanistically impact cancer cell killing or T cell fitness. Finally, we contrast the feasibility and potential benefits of the various approaches using CAR-T and TCE therapeutics and discuss why the promising "IF/THEN" and "NOT" gate types pertain more specifically to CAR-T therapies, but can also succeed by integrating both technologies.
Collapse
Affiliation(s)
| | - Richard Smith
- Cell Biology Research, Kite Pharma, Foster City, CA, United States
| |
Collapse
|
9
|
Zhang SH, Peng LL, Chen YF, Xu Y, Moradi V. Focusing on exosomes to overcome the existing bottlenecks of CAR-T cell therapy. Inflamm Regen 2024; 44:45. [PMID: 39490997 PMCID: PMC11533312 DOI: 10.1186/s41232-024-00358-x] [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: 07/15/2024] [Accepted: 10/22/2024] [Indexed: 11/05/2024] Open
Abstract
Since chimeric antigen receptor T (CAR-T) cells were introduced three decades ago, the treatment using these cells has led to outstanding outcomes, and at the moment, CAR-T cell therapy is a well-established mainstay for treating CD19 + malignancies and multiple myeloma. Despite the astonishing results of CAR-T cell therapy in B-cell-derived malignancies, several bottlenecks must be overcome to promote its safety and efficacy and broaden its applicability. These bottlenecks include cumbersome production process, safety concerns of viral vectors, poor efficacy in treating solid tumors, life-threatening side effects, and dysfunctionality of infused CAR-T cells over time. Exosomes are nano-sized vesicles that are secreted by all living cells and play an essential role in cellular crosstalk by bridging between cells. In this review, we discuss how the existing bottlenecks of CAR-T cell therapy can be overcome by focusing on exosomes. First, we delve into the effect of tumor-derived exosomes on the CAR-T cell function and discuss how inhibiting their secretion can enhance the efficacy of CAR-T cell therapy. Afterward, the application of exosomes to the manufacturing of CAR-T cells in a non-viral approach is discussed. We also review the latest advancements in ex vivo activation and cultivation of CAR-T cells using exosomes, as well as the potential of engineered exosomes to in vivo induction or boost the in vivo proliferation of CAR-T cells. Finally, we discuss how CAR-engineered exosomes can be used as a versatile tool for the direct killing of tumor cells or delivering intended therapeutic payloads in a targeted manner.
Collapse
Affiliation(s)
- Si-Heng Zhang
- Faculty of Medicine, Macau University of Science and Technology, Taipa, Macao SAR, 999078, China
- Zhejiang Provincial Center for Disease Control and Prevention, Hangzhou, Zhejiang, 310000, China
| | - Ling-Long Peng
- Wuhu Hospital, East China Normal University (The Second People's Hospital of Wuhu), Wuhu, 241000, China
| | - Yi-Fei Chen
- Faculty of Medicine, Macau University of Science and Technology, Taipa, Macao SAR, 999078, China
| | - Yan Xu
- Zhejiang Provincial Center for Disease Control and Prevention, Hangzhou, Zhejiang, 310000, China.
| | - Vahid Moradi
- Hematology and Bood Transfusion Science Department, School of Allied Medical Sciences, Tehran University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
10
|
Messaoudi D, Perez F, Gouveia Z. [The new generations of CAR-T cells]. Med Sci (Paris) 2024; 40:848-857. [PMID: 39656982 DOI: 10.1051/medsci/2024151] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2024] Open
Abstract
Chimeric antigen receptor (CAR)-T is one of the most promising modern cancer immunotherapies. In the recent years, impressive results have been obtained in the treatment of cancer which led to FDA approval for the treatment of liquid tumors. In this cell-based therapy, immune cells (e.g. T and NK cells) are engineered to express a synthetic receptor CAR to specifically recognize and eliminate cells expressing a target antigen. CAR has evolved through different generations aiming to boost its biological activity and overcome limitations such as low persistence, limited potency, life-threatening toxicity and inefficient activity against solid tumor. The present review provides an overview of the different CAR generations, starting from the 1st generation with limited cytotoxic activity until the latest generation, the 5th generation or new generation, developed to overcome various limitations of CAR T therapy. The current ongoing clinical trials in cancer and autoimmune diseases, and the limitation associated with CAR-T cells in cancer therapy, are also discussed.
Collapse
Affiliation(s)
- Djamel Messaoudi
- Dynamics of intracellular organization laboratory, Institut Curie, PSL Research University, Sorbonne Université, CNRS, UMR144, Paris, France
| | - Franck Perez
- Dynamics of intracellular organization laboratory, Institut Curie, PSL Research University, Sorbonne Université, CNRS, UMR144, Paris, France
| | - Zélia Gouveia
- Dynamics of intracellular organization laboratory, Institut Curie, PSL Research University, Sorbonne Université, CNRS, UMR144, Paris, France - Cell therapy Acceleration and Innovation (CellAction), Institut Curie, Suresnes, France
| |
Collapse
|
11
|
Garg P, Pareek S, Kulkarni P, Horne D, Salgia R, Singhal SS. Next-Generation Immunotherapy: Advancing Clinical Applications in Cancer Treatment. J Clin Med 2024; 13:6537. [PMID: 39518676 PMCID: PMC11546714 DOI: 10.3390/jcm13216537] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2024] [Revised: 10/28/2024] [Accepted: 10/28/2024] [Indexed: 11/16/2024] Open
Abstract
Next-generation immunotherapies have revolutionized cancer treatment, offering hope for patients with hard-to-treat tumors. This review focuses on the clinical applications and advancements of key immune-based therapies, including immune checkpoint inhibitors, CAR-T cell therapy, and new cancer vaccines designed to harness the immune system to combat malignancies. A prime example is the success of pembrolizumab in the treatment of advanced melanoma, underscoring the transformative impact of these therapies. Combination treatments, integrating immunotherapy with chemotherapy, radiation, and targeted therapies, are demonstrating synergistic benefits and improving patient outcomes. This review also explores the evolving role of personalized immunotherapy, guided by biomarkers, genomic data, and the tumor environment, to better target individual tumors. Although significant progress has been made, challenges such as resistance, side effects, and high treatment costs persist. Technological innovations, including nanotechnology and artificial intelligence, are explored as future enablers of these therapies. The review evaluates key clinical trials, breakthroughs, and the emerging immune-modulating agents and advanced delivery systems that hold great promise for enhancing treatment efficacy, reducing toxicity, and expanding access to immunotherapy. In conclusion, this review highlights the ongoing advancements in immunotherapy that are reshaping cancer care, with future strategies poised to overcome current challenges and further extend therapeutic reach.
Collapse
Affiliation(s)
- Pankaj Garg
- Department of Chemistry, GLA University, Mathura 281406, Uttar Pradesh, India
| | - Siddhika Pareek
- Departments of Medical Oncology & Therapeutics Research, Beckman Research Institute of City of Hope, Comprehensive Cancer Center and National Medical Center, Duarte, CA 91010, USA
| | - Prakash Kulkarni
- Departments of Medical Oncology & Therapeutics Research, Beckman Research Institute of City of Hope, Comprehensive Cancer Center and National Medical Center, Duarte, CA 91010, USA
| | - David Horne
- Departments of Molecular Medicine, Beckman Research Institute of City of Hope, Comprehensive Cancer Center and National Medical Center, Duarte, CA 91010, USA
| | - Ravi Salgia
- Departments of Medical Oncology & Therapeutics Research, Beckman Research Institute of City of Hope, Comprehensive Cancer Center and National Medical Center, Duarte, CA 91010, USA
| | - Sharad S. Singhal
- Departments of Medical Oncology & Therapeutics Research, Beckman Research Institute of City of Hope, Comprehensive Cancer Center and National Medical Center, Duarte, CA 91010, USA
| |
Collapse
|
12
|
Hegazi A, Rager LE, Watkins DE, Su KH. Advancing Immunotherapy in Pancreatic Cancer. Int J Mol Sci 2024; 25:11560. [PMID: 39519112 PMCID: PMC11546161 DOI: 10.3390/ijms252111560] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2024] [Revised: 10/21/2024] [Accepted: 10/22/2024] [Indexed: 11/16/2024] Open
Abstract
Pancreatic cancer remains one of the deadliest malignancies, with a consistently low five-year survival rate for the past several decades. This is in stark contrast to other cancers, which have seen significant improvement in survival and prognosis due to recent developments in therapeutic modalities. These modest improvements in pancreatic cancer outcomes have primarily resulted from minor advances in cytotoxic chemotherapeutics, with limited progress in other treatment approaches. A major focus of current therapeutic research is the further development of immunomodulatory therapies characterized by antibody-based approaches, cellular therapies, and vaccines. Although initial results utilizing immunotherapy in pancreatic cancer have been mixed, recent clinical trials have demonstrated significant improvements in patient outcomes. In this review, we detail these three approaches to immunomodulation, highlighting their common targets and distinct shortcomings, and we provide a narrative summary of completed and ongoing clinical trials that utilize these approaches to immunomodulation. Within this context, we aim to inform future research efforts by identifying promising areas that warrant further exploration.
Collapse
Affiliation(s)
| | | | | | - Kuo-Hui Su
- Department of Cell and Cancer Biology, College of Medicine and Life Sciences, The University of Toledo, Toledo, OH 43614, USA; (A.H.); (L.E.R.); (D.E.W.)
| |
Collapse
|
13
|
Masnikosa R, Cvetković Z, Pirić D. Tumor Biology Hides Novel Therapeutic Approaches to Diffuse Large B-Cell Lymphoma: A Narrative Review. Int J Mol Sci 2024; 25:11384. [PMID: 39518937 PMCID: PMC11545713 DOI: 10.3390/ijms252111384] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2024] [Revised: 10/13/2024] [Accepted: 10/16/2024] [Indexed: 11/16/2024] Open
Abstract
Diffuse large B-cell lymphoma (DLBCL) is a malignancy of immense biological and clinical heterogeneity. Based on the transcriptomic or genomic approach, several different classification schemes have evolved over the years to subdivide DLBCL into clinically (prognostically) relevant subsets, but each leaves unclassified samples. Herein, we outline the DLBCL tumor biology behind the actual and potential drug targets and address the challenges and drawbacks coupled with their (potential) use. Therapeutic modalities are discussed, including small-molecule inhibitors, naked antibodies, antibody-drug conjugates, chimeric antigen receptors, bispecific antibodies and T-cell engagers, and immune checkpoint inhibitors. Candidate drugs explored in ongoing clinical trials are coupled with diverse toxicity issues and refractoriness to drugs. According to the literature on DLBCL, the promise for new therapeutic targets lies in epigenetic alterations, B-cell receptor and NF-κB pathways. Herein, we present putative targets hiding in lipid pathways, ferroptosis, and the gut microbiome that could be used in addition to immuno-chemotherapy to improve the general health status of DLBCL patients, thus increasing the chance of being cured. It may be time to devote more effort to exploring DLBCL metabolism to discover novel druggable targets. We also performed a bibliometric and knowledge-map analysis of the literature on DLBCL published from 2014-2023.
Collapse
Affiliation(s)
- Romana Masnikosa
- Department of Physical Chemistry, Vinca Institute of Nuclear Sciences—National Institute of the Republic of Serbia, University of Belgrade, Mike Petrovica Alasa 12-14, 11000 Belgrade, Serbia;
| | - Zorica Cvetković
- Department of Hematology, Clinical Hospital Centre Zemun, Vukova 9, 11000 Belgrade, Serbia
- Faculty of Medicine, University of Belgrade, Dr Subotića 8, 11000 Belgrade, Serbia
| | - David Pirić
- Department of Physical Chemistry, Vinca Institute of Nuclear Sciences—National Institute of the Republic of Serbia, University of Belgrade, Mike Petrovica Alasa 12-14, 11000 Belgrade, Serbia;
| |
Collapse
|
14
|
Aizaz M, Khan AS, Khan M, Musazade E, Yang G. Advancements in tumor-infiltrating lymphocytes: Historical insights, contemporary milestones, and future directions in oncology therapy. Crit Rev Oncol Hematol 2024; 202:104471. [PMID: 39117163 DOI: 10.1016/j.critrevonc.2024.104471] [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: 08/31/2023] [Revised: 07/28/2024] [Accepted: 08/02/2024] [Indexed: 08/10/2024] Open
Abstract
Tumor-infiltrating lymphocytes (TILs) are a subtype of immune cells that infiltrate and accumulate within tumors. Studies proved that TILs can be used as prognostic and predictive markers for cancer patients' responses to immunotherapy. This review explores the modern knowledge of TILs, the challenges and opportunities for utilizing TILs in cancer treatment, such as the rise of therapies under TIL circumstances, the identification of biomarkers for TIL activity, and methods used to isolate and expand TILs for therapeutic use. Ongoing clinical trials and promising results in different cancer types are highlighted, including melanoma, ovarian, and colorectal cancer. This also focuses on ongoing efforts to improve TIL-based therapies by identifying the specific subsets of TILs that are most effective in treating cancer and developing methods to increase the functionality and persistence of TILs in the tumor microenvironment. The article recapitulates the present state TILs therapy, ongoing research, and improvements to its potency.
Collapse
Affiliation(s)
- Muhammad Aizaz
- Shandong Provincial Key Laboratory of Animal Resistance Biology, College of Life Science, Shandong Normal University, Jinan, China.
| | | | - Maria Khan
- Center of Biotechnology and Microbiology, University of Peshawar, Pakistan.
| | - Elshan Musazade
- College of Life Science, Jilin Agricultural University, Changchun, China.
| | - Guiwen Yang
- Shandong Provincial Key Laboratory of Animal Resistance Biology, College of Life Science, Shandong Normal University, Jinan, China.
| |
Collapse
|
15
|
Lin H, Yang X, Ye S, Huang L, Mu W. Antigen escape in CAR-T cell therapy: Mechanisms and overcoming strategies. Biomed Pharmacother 2024; 178:117252. [PMID: 39098176 DOI: 10.1016/j.biopha.2024.117252] [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: 05/13/2024] [Revised: 07/30/2024] [Accepted: 07/31/2024] [Indexed: 08/06/2024] Open
Abstract
Chimeric antigen receptor T (CAR-T) cell therapy has shown promise in treating hematological malignancies and certain solid tumors. However, its efficacy is often hindered by negative relapses resulting from antigen escape. This review firstly elucidates the mechanisms underlying antigen escape during CAR-T cell therapy, including the enrichment of pre-existing target-negative tumor clones, antigen gene mutations or alternative splicing, deficits in antigen processing, antigen redistribution, lineage switch, epitope masking, and trogocytosis-mediated antigen loss. Furthermore, we summarize various strategies to overcome antigen escape, evaluate their advantages and limitations, and propose future research directions. Thus, we aim to provide valuable insights to enhance the effectiveness of CAR-T cell therapy.
Collapse
Affiliation(s)
- Haolong Lin
- Department of Hematology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430030, China; Immunotherapy Research Center for Hematologic Diseases of Hubei Province, Wuhan, Hubei 430030, China
| | - Xiuxiu Yang
- Department of Hematology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430030, China; Immunotherapy Research Center for Hematologic Diseases of Hubei Province, Wuhan, Hubei 430030, China
| | - Shanwei Ye
- Department of Hematology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430030, China; Immunotherapy Research Center for Hematologic Diseases of Hubei Province, Wuhan, Hubei 430030, China
| | - Liang Huang
- Department of Hematology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430030, China; Immunotherapy Research Center for Hematologic Diseases of Hubei Province, Wuhan, Hubei 430030, China; State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Haihe Laboratory of Cell Ecosystem, Institute of Hematology & Blood Diseases Hospital, Chinese Academy of Medical Science & Peking Union Medical College, Tianjin 300020, China; Tianjin Institutes of Health Science, Tianjin 301600, China.
| | - Wei Mu
- Department of Hematology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430030, China; Immunotherapy Research Center for Hematologic Diseases of Hubei Province, Wuhan, Hubei 430030, China.
| |
Collapse
|
16
|
Bar O, Porgador A, Cooks T. Exploring the potential of the convergence between extracellular vesicles and CAR technology as a novel immunotherapy approach. JOURNAL OF EXTRACELLULAR BIOLOGY 2024; 3:e70011. [PMID: 39328262 PMCID: PMC11424882 DOI: 10.1002/jex2.70011] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 03/15/2024] [Revised: 08/20/2024] [Accepted: 09/04/2024] [Indexed: 09/28/2024]
Abstract
Cancer therapy is a dynamically evolving field, witnessing the emergence of innovative approaches that offer a promising outlook for patients grappling with persistent disease. Within the realm of therapeutic exploration, chimeric antigen receptor (CAR) T cells as well as CAR NK cells, have surfaced as novel approaches, each possessing unique attributes and transformative potential. Immune cells engineered to express CARs recognizing tumour-specific antigens, have shown remarkable promise in treating terminal cancers by combining the precision of antibody specificity with the potent cytotoxic function of T cells. However, their application in solid tumours is still in its nascent stages, presenting unique major challenges. On the same note, CAR NK cells offer a distinct immunotherapeutic approach, utilizing CARs on NK cells, providing advantages in safety, manufacturing simplicity, and a broader scope for cancer treatment. Extracellular vesicles (EVs) have emerged as promising therapeutic agents due to their ability to carry crucial biomarkers and biologically active molecules, serving as vital messengers in the intercellular communication network. In the context of cancer, the therapeutic potential of EVs lies in delivering tumour-suppressing proteins, nucleic acid components, or targeting drugs with precision, thereby redefining the paradigm of precision medicine. The fusion of CAR technology with the capabilities of EVs has given rise to a new therapeutic frontier. CAR T EVs and CAR NK EVs, leveraging the power of EVs, have the potential to alleviate challenges associated with live-cell therapies. EVs are suggested to reduce the side effects linked to CAR T cell therapy and hold the potential to revolutionize the penetrance in solid tumours. EVs act as carriers of pro-apoptotic molecules and RNA components, enhancing immune responses and thereby expanding their therapeutic potential. In this review article, we navigate dynamic landscapes, with our objective being to evaluate comparative efficacy, safety profiles, manufacturing complexities, and clinical applicability.
Collapse
Affiliation(s)
- Ofir Bar
- The Shraga Segal Department of Microbiology, Immunology and Genetics, Faculty of Health Sciences Ben-Gurion University Beer-Sheva Israel
| | - Angel Porgador
- The Shraga Segal Department of Microbiology, Immunology and Genetics, Faculty of Health Sciences Ben-Gurion University Beer-Sheva Israel
| | - Tomer Cooks
- The Shraga Segal Department of Microbiology, Immunology and Genetics, Faculty of Health Sciences Ben-Gurion University Beer-Sheva Israel
| |
Collapse
|
17
|
Li W, Wang X, Zhang X, Aziz AUR, Wang D. CAR-NK Cell Therapy: A Transformative Approach to Overcoming Oncological Challenges. Biomolecules 2024; 14:1035. [PMID: 39199421 PMCID: PMC11352442 DOI: 10.3390/biom14081035] [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: 06/14/2024] [Revised: 07/27/2024] [Accepted: 08/13/2024] [Indexed: 09/01/2024] Open
Abstract
The use of chimeric antigen receptor (CAR) in natural killer (NK) cells for cancer therapy is gaining momentum, marking a significant shift in cancer treatment. This review aims to explore the potential of CAR-NK cell therapy in cancer immunotherapy, providing a fresh perspective. It discusses the innovative approaches in CAR-NK cell design and engineering, particularly targeting refractory or recurrent cancers. By comparing CAR-NK cells with traditional therapies, the review highlights their unique ability to tackle tumor heterogeneity and immune system suppression. Additionally, it explains how novel cytokines and receptors can enhance CAR-NK cell efficacy, specificity, and functionality. This review underscores the advantages of CAR-NK cells, including reduced toxicity, lower cost, and broader accessibility compared to CAR-T cells, along with their potential in treating both blood cancers and solid tumors.
Collapse
Affiliation(s)
- Wangshu Li
- China Key Laboratory for Early Diagnosis and Biotherapy of Malignant Tumors in Children and Women, Dalian Women and Children’s Medical Group, Dalian 116012, China; (W.L.); (X.W.)
- Department of Obstetrics and Gynecology, Shengjing Hospital of China Medical University, Shenyang 110004, China
| | - Xiuying Wang
- China Key Laboratory for Early Diagnosis and Biotherapy of Malignant Tumors in Children and Women, Dalian Women and Children’s Medical Group, Dalian 116012, China; (W.L.); (X.W.)
| | - Xu Zhang
- The Second Affiliated Hospital of Harbin Medical University, Harbin 151801, China;
| | - Aziz ur Rehman Aziz
- China Key Laboratory for Early Diagnosis and Biotherapy of Malignant Tumors in Children and Women, Dalian Women and Children’s Medical Group, Dalian 116012, China; (W.L.); (X.W.)
| | - Daqing Wang
- China Key Laboratory for Early Diagnosis and Biotherapy of Malignant Tumors in Children and Women, Dalian Women and Children’s Medical Group, Dalian 116012, China; (W.L.); (X.W.)
| |
Collapse
|
18
|
Tyrna P, Procyk G, Szeleszczuk Ł, Młynarczuk-Biały I. Different Strategies to Overcome Resistance to Proteasome Inhibitors-A Summary 20 Years after Their Introduction. Int J Mol Sci 2024; 25:8949. [PMID: 39201634 PMCID: PMC11354503 DOI: 10.3390/ijms25168949] [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: 07/02/2024] [Revised: 08/09/2024] [Accepted: 08/14/2024] [Indexed: 09/02/2024] Open
Abstract
Proteasome inhibitors (PIs), bortezomib, carfilzomib, and ixazomib, are the first-line treatment for multiple myeloma (MM). They inhibit cytosolic protein degradation in cells, which leads to the accumulation of misfolded and malfunctioned proteins in the cytosol and endoplasmic reticulum, resulting in cell death. Despite being a breakthrough in MM therapy, malignant cells develop resistance to PIs via different mechanisms. Understanding these mechanisms drives research toward new anticancer agents to overcome PI resistance. In this review, we summarize the mechanism of action of PIs and how MM cells adapt to these drugs to develop resistance. Finally, we explore these mechanisms to present strategies to interfere with PI resistance. The strategies include new inhibitors of the ubiquitin-proteasome system, drug efflux inhibitors, autophagy disruption, targeting stress response mechanisms, affecting survival and cell cycle regulators, bone marrow microenvironment modulation, and immunotherapy. We list potential pharmacological targets examined in in vitro, in vivo, and clinical studies. Some of these strategies have already provided clinicians with new anti-MM medications, such as panobinostat and selinexor. We hope that further exploration of the subject will broaden the range of therapeutic options and improve patient outcomes.
Collapse
Affiliation(s)
- Paweł Tyrna
- Histology and Embryology Students’ Science Association, Department of Histology and Embryology, Faculty of Medicine, Warsaw Medical University, Chalubinskiego 5, 02-004 Warsaw, Poland;
| | - Grzegorz Procyk
- 1st Chair and Department of Cardiology, Medical University of Warsaw, Banacha 1A, 02-097 Warsaw, Poland;
| | - Łukasz Szeleszczuk
- Department of Organic and Physical Chemistry, Faculty of Pharmacy, Medical University of Warsaw, Banacha 1 Str., 02-093 Warsaw, Poland;
| | - Izabela Młynarczuk-Biały
- Department of Histology and Embryology, Faculty of Medicine, Warsaw Medical University, Chalubinskiego 5, 02-004 Warsaw, Poland
| |
Collapse
|
19
|
Zarychta J, Kowalczyk A, Marszołek A, Zawitkowska J, Lejman M. Strategies to overcome tumor microenvironment immunosuppressive effect on the functioning of CAR-T cells in high-grade glioma. Ther Adv Med Oncol 2024; 16:17588359241266140. [PMID: 39156126 PMCID: PMC11327996 DOI: 10.1177/17588359241266140] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2024] [Accepted: 06/18/2024] [Indexed: 08/20/2024] Open
Abstract
Despite significant progress in the treatment of some types of cancer, high-grade gliomas (HGGs) remain a significant clinical problem. In the case of glioblastoma (GBM), the most common solid tumor of the central nervous system in adults, the average survival time from diagnosis is only 15-18 months, despite the use of intensive multimodal therapy. Chimeric antigen receptor (CAR)-expressing T cells, which have already been approved by the Food and Drug Administration for use in the treatment of certain hematologic malignancies, are a new, promising therapeutic option. However, the efficacy of CAR-T cells in solid tumors is lower due to the immunosuppressive tumor microenvironment (TME). Reprogramming the immunosuppressive TME toward a pro-inflammatory phenotype therefore seems particularly important because it may allow for increasing the effectiveness of CAR-T cells in the therapy of solid tumors. The following literature review aims to present the results of preclinical studies showing the possibilities of improving the efficacy of CAR-T in the TME of GBM by reprogramming the TME toward a pro-inflammatory phenotype. It may be achievable thanks to the use of CAR-T in a synergistic therapy in combination with oncolytic viruses, radiotherapy, or epigenetic inhibitors, as well as by supporting CAR-T cells crossing of the blood-brain barrier, normalizing impaired angiogenesis in the TME, improving CAR-T effector functions by cytokine signaling or by blocking/knocking out T-cell inhibitors, and modulating the microRNA expression. The use of CAR-T cells modified in this way in synergistic therapy could lead to the longer survival of patients with HGG by inducing an endogenous anti-tumor response.
Collapse
Affiliation(s)
- Julia Zarychta
- Student Scientific Society of Department of Pediatric Hematology, Oncology and Transplantology, Medical University of Lublin, Lublin, Poland
| | - Adrian Kowalczyk
- Student Scientific Society of Department of Pediatric Hematology, Oncology and Transplantology, Medical University of Lublin, Lublin, Poland
| | - Anna Marszołek
- Student Scientific Society of Independent Laboratory of Genetic Diagnostics, Medical University of Lublin, Lublin, Poland
| | - Joanna Zawitkowska
- Department of Pediatric Hematology, Oncology and Transplantology, Medical University of Lublin, Lublin, Poland
| | - Monika Lejman
- Independent Laboratory of Genetic Diagnostics, Medical University of Lublin, A. Racławickie 1, Lublin 20-093, Poland
| |
Collapse
|
20
|
Duan R, Milton P, Sittplangkoon C, Liu X, Sui Z, Boyce BF, Yao Z. Chimeric antigen receptor dendritic cells targeted delivery of a single tumoricidal factor for cancer immunotherapy. Cancer Immunol Immunother 2024; 73:203. [PMID: 39105847 PMCID: PMC11303651 DOI: 10.1007/s00262-024-03788-1] [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: 02/23/2024] [Accepted: 07/19/2024] [Indexed: 08/07/2024]
Abstract
BACKGROUND Chimeric antigen receptor (CAR)-T cells have been used to treat blood cancers by producing a wide variety of cytokines. However, they are not effective in treating solid cancers and can cause severe side-effects, including cytokine release syndrome. TNFα is a tumoricidal cytokine, but it markedly increases the protein levels of cIAP1 and cIAP2, the members of inhibitor of apoptosis protein (IAP) family of E3 ubiquitin ligase that limits caspase-induced apoptosis. Degradation of IAP proteins by an IAP antagonist does not effectively kill cancer cells but enables TNFα to strongly induce cancer cell apoptosis. It would be a promising approach to treat cancers by targeted delivery of TNFα through an inactive adoptive cell in combination with an IAP antagonist. METHODS Human dendritic cells (DCs) were engineered to express a single tumoricidal factor, TNFα, and a membrane-anchored Mucin1 antibody scFv, named Mucin 1 directed DCs expressing TNFα (M-DCsTNF). The efficacy of M-DCsTNF in recognizing and treating breast cancer was tested in vitro and in vivo. RESULTS Mucin1 was highly expressed on the surface of a wide range of human breast cancer cell lines. M-DCsTNF directly associated with MDA-MB-231 cells in the bone of NSG mice. M-DCsTNF plus an IAP antagonist, SM-164, but neither alone, markedly induce MDA-MB-231 breast cancer cell apoptosis, which was blocked by TNF antibody. Importantly, M-DCsTNF combined with SM-164, but not SM-164 alone, inhibited the growth of patient-derived breast cancer in NSG mice. CONCLUSION An adoptive cell targeting delivery of TNFα combined with an IAP antagonist is a novel effective approach to treat breast cancer and could be expanded to treat other solid cancers. Unlike CAR-T cell, this novel adoptive cell is not activated to produce a wide variety of cytokines, except for additional overexpressed TNF, and thus could avoid the severe side effects such as cytokine release syndrome.
Collapse
Affiliation(s)
- Rong Duan
- Department of Pathology and Laboratory Medicine, University of Rochester Medical Center, 601 Elmwood Ave, Rochester, NY, 14642, USA
| | - Philip Milton
- Department of Pathology and Laboratory Medicine, University of Rochester Medical Center, 601 Elmwood Ave, Rochester, NY, 14642, USA
- School of Engineering, University of Rochester, Rochester, NY, 14627, USA
| | - Chutamath Sittplangkoon
- Department of Pathology and Laboratory Medicine, University of Rochester Medical Center, 601 Elmwood Ave, Rochester, NY, 14642, USA
| | - Xin Liu
- Department of Pathology and Laboratory Medicine, University of Rochester Medical Center, 601 Elmwood Ave, Rochester, NY, 14642, USA
- Department of Orthopedics, Tianjin Hospital, Tianjin, 30021, People's Republic of China
| | - Zhining Sui
- Department of Biostatistics and Computational Biology, University of Rochester Medical Center, 601 Elmwood Ave, Rochester, NY, 14642, USA
| | - Brendan F Boyce
- Department of Pathology and Laboratory Medicine, University of Rochester Medical Center, 601 Elmwood Ave, Rochester, NY, 14642, USA
| | - Zhenqiang Yao
- Department of Pathology and Laboratory Medicine, University of Rochester Medical Center, 601 Elmwood Ave, Rochester, NY, 14642, USA.
| |
Collapse
|
21
|
Wang M, Jia L, Dai X, Zhang X. Advanced strategies in improving the immunotherapeutic effect of CAR-T cell therapy. Mol Oncol 2024; 18:1821-1848. [PMID: 38456710 PMCID: PMC11306536 DOI: 10.1002/1878-0261.13621] [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: 08/12/2023] [Revised: 12/23/2023] [Accepted: 02/20/2024] [Indexed: 03/09/2024] Open
Abstract
Chimeric antigen receptor (CAR-T) cell therapy is a newly developed immunotherapy strategy and has achieved satisfactory outcomes in the treatment of hematological malignancies. However, some adverse effects related to CAR-T cell therapy have to be resolved before it is widely used in clinics as a cancer treatment. Furthermore, the application of CAR-T cell therapy in the treatment of solid tumors has been hampered by numerous limitations. Therefore, it is essential to explore novel strategies to improve the therapeutic effect of CAR-T cell therapy. In this review, we summarized the recently developed strategies aimed at optimizing the generation of CAR-T cells and improving the anti-tumor efficiency of CAR-T cell therapy. Furthermore, the discovery of new targets for CAR-T cell therapy and the combined treatment strategies of CAR-T cell therapy with chemotherapy, radiotherapy, cancer vaccines and nanomaterials are highlighted.
Collapse
Affiliation(s)
- Minmin Wang
- Key Laboratory of Organ Regeneration and Transplantation of Ministry of EducationFirst Hospital of Jilin UniversityChangchunChina
- National‐Local Joint Engineering Laboratory of Animal Models for Human DiseaseFirst Hospital of Jilin UniversityChangchunChina
| | - Linzi Jia
- Department of General MedicineShanxi Province Cancer HospitalTaiyuanChina
| | - Xiangpeng Dai
- Key Laboratory of Organ Regeneration and Transplantation of Ministry of EducationFirst Hospital of Jilin UniversityChangchunChina
- National‐Local Joint Engineering Laboratory of Animal Models for Human DiseaseFirst Hospital of Jilin UniversityChangchunChina
| | - Xiaoling Zhang
- Key Laboratory of Organ Regeneration and Transplantation of Ministry of EducationFirst Hospital of Jilin UniversityChangchunChina
- National‐Local Joint Engineering Laboratory of Animal Models for Human DiseaseFirst Hospital of Jilin UniversityChangchunChina
| |
Collapse
|
22
|
Ohno R, Nakamura A. Advancing autoimmune Rheumatic disease treatment: CAR-T Cell Therapies - Evidence, Safety, and future directions. Semin Arthritis Rheum 2024; 67:152479. [PMID: 38810569 DOI: 10.1016/j.semarthrit.2024.152479] [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: 03/09/2024] [Revised: 04/20/2024] [Accepted: 05/08/2024] [Indexed: 05/31/2024]
Abstract
INTRODUCTION Despite advancements in managing autoimmune rheumatic diseases (ARDs) with existing treatments, many patients still encounter challenges such as inadequate responses, difficulty in maintaining remission, and side effects. Chimeric Antigen Receptor (CAR) T-cell therapy, originally developed for cancer, has now emerged as a promising option for cases of refractory ARDs. METHODS A search of the literature was conducted to compose a narrative review exploring the current evidence, potential safety, limitations, potential modifications, and future directions of CAR-T cells in ARDs. RESULTS CAR-T cell therapy has been administered to patients with refractory ARDs, including systemic lupus erythematosus, antisynthetase syndrome, and systemic sclerosis, demonstrating significant improvement. Notable responses include enhanced clinical symptoms, reduced serum autoantibody titers, and sustained remissions in disease activity. Preclinical and in vitro studies using both animal and human samples also support the efficacy and elaborate on potential mechanisms of CAR-T cells against antineutrophil cytoplasmic antibody-associated vasculitis and rheumatoid arthritis. While cautious monitoring of adverse events, such as cytokine release syndrome, is crucial, the therapy appears to be highly tolerable. Nevertheless, challenges persist, including cost, durability due to potential CAR-T cell exhaustion, and manufacturing complexities, urging the development of innovative solutions to further enhance CAR-T cell therapy accessibility in ARDs. CONCLUSIONS CAR-T cell therapy for refractory ARDs has demonstrated high effectiveness. While no significant warning signs are currently reported, achieving a balance between therapeutic efficacy and safety is vital in adapting CAR-T cell therapy for ARDs. Moreover, there is significant potential for technological advancements to enhance the delivery of this treatment to patients, thereby ensuring safer and more effective disease control for patients.
Collapse
Affiliation(s)
- Ryunosuke Ohno
- Department of Medicine, Division of Rheumatology, Queen's University, Kingston, Ontario, Canada; Department of Medicine, Okayama University, Okayama, Japan
| | - Akihiro Nakamura
- Department of Medicine, Division of Rheumatology, Queen's University, Kingston, Ontario, Canada; Translational Institute of Medicine, School of Medicine, Queen's University, Ontario, Canada; Rheumatology Clinic, Kingston Health Science Centre, Kingston, Ontario, Canada.
| |
Collapse
|
23
|
De Castro V, Galaine J, Loyon R, Godet Y. CRISPR-Cas gene knockouts to optimize engineered T cells for cancer immunotherapy. Cancer Gene Ther 2024; 31:1124-1134. [PMID: 38609574 DOI: 10.1038/s41417-024-00771-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2024] [Revised: 04/02/2024] [Accepted: 04/03/2024] [Indexed: 04/14/2024]
Abstract
While CAR-T and tgTCR-T therapies have exhibited noteworthy and promising outcomes in hematologic and solid tumors respectively, a set of distinct challenges remains. Consequently, the quest for novel strategies has become imperative to safeguard and more effectively release the full functions of engineered T cells. These factors are intricately linked to the success of adoptive cell therapy. Recently, CRISPR-based technologies have emerged as a major breakthrough for maintaining T cell functions. These technologies have allowed the discovery of T cells' negative regulators such as specific cell-surface receptors, cell-signaling proteins, and transcription factors that are involved in the development or maintenance of T cell dysfunction. By employing a CRISPR-genic invalidation approach to target these negative regulators, it has become possible to prevent the emergence of hypofunctional T cells. This review revisits the establishment of the dysfunctional profile of T cells before delving into a comprehensive summary of recent CRISPR-gene invalidations, with each invalidation contributing to the enhancement of engineered T cells' antitumor capacities. The narrative unfolds as we explore how these advancements were discovered and identified, marking a significant advancement in the pursuit of superior adoptive cell therapy.
Collapse
Affiliation(s)
- Valentine De Castro
- Université de Franche-Comté, EFS, INSERM, UMR RIGHT, F-25000, Besançon, France
| | - Jeanne Galaine
- Université de Franche-Comté, EFS, INSERM, UMR RIGHT, F-25000, Besançon, France
| | - Romain Loyon
- Université de Franche-Comté, EFS, INSERM, UMR RIGHT, F-25000, Besançon, France
| | - Yann Godet
- Université de Franche-Comté, EFS, INSERM, UMR RIGHT, F-25000, Besançon, France.
| |
Collapse
|
24
|
Olejarz W, Sadowski K, Szulczyk D, Basak G. Advancements in Personalized CAR-T Therapy: Comprehensive Overview of Biomarkers and Therapeutic Targets in Hematological Malignancies. Int J Mol Sci 2024; 25:7743. [PMID: 39062986 PMCID: PMC11276786 DOI: 10.3390/ijms25147743] [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: 06/30/2024] [Revised: 07/12/2024] [Accepted: 07/12/2024] [Indexed: 07/28/2024] Open
Abstract
Chimeric antigen receptor T-cell (CAR-T) therapy is a novel anticancer therapy using autologous or allogeneic T-cells. To date, six CAR-T therapies for specific B-cell acute lymphoblastic leukemia (B-ALL), non-Hodgkin lymphomas (NHL), and multiple myeloma (MM) have been approved by the Food and Drug Administration (FDA). Significant barriers to the effectiveness of CAR-T therapy include cytokine release syndrome (CRS), neurotoxicity in the case of Allogeneic Stem Cell Transplantation (Allo-SCT) graft-versus-host-disease (GVHD), antigen escape, modest antitumor activity, restricted trafficking, limited persistence, the immunosuppressive microenvironment, and senescence and exhaustion of CAR-Ts. Furthermore, cancer drug resistance remains a major problem in clinical practice. CAR-T therapy, in combination with checkpoint blockades and bispecific T-cell engagers (BiTEs) or other drugs, appears to be an appealing anticancer strategy. Many of these agents have shown impressive results, combining efficacy with tolerability. Biomarkers like extracellular vesicles (EVs), cell-free DNA (cfDNA), circulating tumor (ctDNA) and miRNAs may play an important role in toxicity, relapse assessment, and efficacy prediction, and can be implicated in clinical applications of CAR-T therapy and in establishing safe and efficacious personalized medicine. However, further research is required to fully comprehend the particular side effects of immunomodulation, to ascertain the best order and combination of this medication with conventional chemotherapy and targeted therapies, and to find reliable predictive biomarkers.
Collapse
Affiliation(s)
- Wioletta Olejarz
- Department of Biochemistry and Pharmacogenomics, Faculty of Pharmacy, Medical University of Warsaw, 02-097 Warsaw, Poland;
- Centre for Preclinical Research, Medical University of Warsaw, 02-097 Warsaw, Poland
| | - Karol Sadowski
- Department of Biochemistry and Pharmacogenomics, Faculty of Pharmacy, Medical University of Warsaw, 02-097 Warsaw, Poland;
- Centre for Preclinical Research, Medical University of Warsaw, 02-097 Warsaw, Poland
- Department of Hematology, Transplantation and Internal Medicine, Medical University of Warsaw, 02-097 Warsaw, Poland;
| | - Daniel Szulczyk
- Chair and Department of Biochemistry, The Medical University of Warsaw, 02-097 Warsaw, Poland;
| | - Grzegorz Basak
- Department of Hematology, Transplantation and Internal Medicine, Medical University of Warsaw, 02-097 Warsaw, Poland;
| |
Collapse
|
25
|
Elsayed A, Plüss L, Nideroest L, Rotta G, Thoma M, Zangger N, Peissert F, Pfister SK, Pellegrino C, Dakhel Plaza S, De Luca R, Manz MG, Oxenius A, Puca E, Halin C, Neri D. Optimizing the Design and Geometry of T Cell-Engaging Bispecific Antibodies Targeting CEA in Colorectal Cancer. Mol Cancer Ther 2024; 23:1010-1020. [PMID: 38638035 DOI: 10.1158/1535-7163.mct-23-0766] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2023] [Revised: 03/05/2024] [Accepted: 04/05/2024] [Indexed: 04/20/2024]
Abstract
Metastatic colorectal cancer remains a leading cause of cancer-related deaths, with a 5-year survival rate of only 15%. T cell-engaging bispecific antibodies (TCBs) represent a class of biopharmaceuticals that redirect cytotoxic T cells toward tumor cells, thereby turning immunologically "cold" tumors into "hot" ones. The carcinoembryonic antigen (CEA) is an attractive tumor-associated antigen that is overexpressed in more than 98% of patients with colorectal cancer. In this study, we report the comparison of four different TCB formats employing the antibodies F4 (targeting human CEA) and 2C11 (targeting mouse CD3ε). These formats include both antibody fragment-based and IgG-based constructs, with either one or two binding specificities of the respective antibodies. The 2 + 1 arrangement, using an anti-CEA single-chain diabody fused to an anti-CD3 single-chain variable fragment, emerged as the most potent design, showing tumor killing at subnanomolar concentrations across three different CEA+ cell lines. The in vitro activity was three times greater in C57BL/6 mouse colon adenocarcinoma cells (MC38) expressing high levels of CEA compared with those expressing low levels, highlighting the impact of CEA density in this assay. The optimal TCB candidate was tested in two different immunocompetent mouse models of colorectal cancer and showed tumor growth retardation. Ex vivo analysis of tumor infiltrates showed an increase in CD4+ and CD8+ T cells upon TCB treatment. This study suggests that bivalent tumor targeting, monovalent T-cell targeting, and a short spatial separation are promising characteristics for CEA-targeting TCBs.
Collapse
Affiliation(s)
- Abdullah Elsayed
- Philochem AG, Otelfingen, Switzerland
- Swiss Federal Institute of Technology (ETH Zürich), Zurich, Switzerland
| | - Louis Plüss
- Philochem AG, Otelfingen, Switzerland
- Swiss Federal Institute of Technology (ETH Zürich), Zurich, Switzerland
| | - Larissa Nideroest
- Swiss Federal Institute of Technology (ETH Zürich), Zurich, Switzerland
| | | | - Marina Thoma
- Swiss Federal Institute of Technology (ETH Zürich), Zurich, Switzerland
| | - Nathan Zangger
- Swiss Federal Institute of Technology (ETH Zürich), Zurich, Switzerland
| | | | | | - Christian Pellegrino
- Department of Medical Oncology and Hematology, University Hospital Zurich and University of Zurich, Comprehensive Cancer Center Zurich (CCCZ), Zurich, Switzerland
| | | | | | - Markus G Manz
- Department of Medical Oncology and Hematology, University Hospital Zurich and University of Zurich, Comprehensive Cancer Center Zurich (CCCZ), Zurich, Switzerland
| | - Annette Oxenius
- Swiss Federal Institute of Technology (ETH Zürich), Zurich, Switzerland
| | | | - Cornelia Halin
- Swiss Federal Institute of Technology (ETH Zürich), Zurich, Switzerland
| | - Dario Neri
- Philochem AG, Otelfingen, Switzerland
- Swiss Federal Institute of Technology (ETH Zürich), Zurich, Switzerland
- Philogen SpA, Siena, Italy
| |
Collapse
|
26
|
Stewart MD, Kalos M, Coutinho V, Better M, Jazayeri J, Yohrling J, Jadlowsky J, Fuchs M, Gidwani S, Goessl C, Hanley PJ, Healy J, Liu W, McKelvey BA, Pearce L, Pilon-Thomas S, Andrews HS, Veldman M, Vong J, Weinbach SP, Allen JD. Accelerating the development of genetically engineered cellular therapies: a framework for extrapolating data across related products. Cytotherapy 2024; 26:778-784. [PMID: 38583170 DOI: 10.1016/j.jcyt.2024.03.009] [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: 10/23/2023] [Revised: 03/11/2024] [Accepted: 03/11/2024] [Indexed: 04/09/2024]
Abstract
BACKGROUND Significant advancements have been made in the field of cellular therapy as anti-cancer treatments, with the approval of chimeric antigen receptor (CAR)-T cell therapies and the development of other genetically engineered cellular therapies. CAR-T cell therapies have demonstrated remarkable clinical outcomes in various hematological malignancies, establishing their potential to change the current cancer treatment paradigm. Due to the increasing importance of genetically engineered cellular therapies in the oncology treatment landscape, implementing strategies to expedite development and evidence generation for the next generation of cellular therapy products can have a positive impact on patients. METHODS We outline a risk-based methodology and assessment aid for the data extrapolation approach across related genetically engineered cellular therapy products. This systematic data extrapolation approach has applicability beyond CAR-T cells and can influence clinical development strategies for a variety of immune therapies such as T cell receptor (TCR) or genetically engineered and other cell-based therapies (e.g., tumor infiltrating lymphocytes, natural killer cells and macrophages). RESULTS By analyzing commonalities in manufacturing processes, clinical trial designs, and regulatory considerations, key learnings were identified. These insights support optimization of the development and regulatory approval of novel cellular therapies. CONCLUSIONS The field of cellular therapy holds immense promise in safely and effectively treating cancer. The ability to extrapolate data across related products presents opportunities to streamline the development process and accelerate the delivery of novel therapies to patients.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Julie Jadlowsky
- Center for Cellular Immunotherapies, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | | | | | | | - Patrick J Hanley
- Center for Cancer and Immunology Research, Children's National Hospital, The George Washington University, Washington, DC, USA
| | | | - Wen Liu
- Lyell Immunopharma, South San Francisco, CA, USA
| | | | - Laura Pearce
- Canadian Cancer Trials Group, Queens University, Kingston, Ontario, Canada
| | | | | | | | | | | | | |
Collapse
|
27
|
Ramoni D, Montecucco F, Carbone F. CAR T therapy from haematological malignancies to aging-related diseases: An ever-expanding universe. Eur J Clin Invest 2024; 54:e14203. [PMID: 38551245 DOI: 10.1111/eci.14203] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/28/2024] [Revised: 03/19/2024] [Accepted: 03/21/2024] [Indexed: 06/06/2024]
Abstract
BACKGROUND Short but impactful, the two-decade story of gene editing allowed a significant breakthrough in the treatment of haematological malignancies. However, despite different generations of chimeric antigen receptor T (CAR T), such a successful therapy has not yet been replicated in solid tumours and non-oncological diseases. METHODS This narrative review discusses how CAR T therapy still faces challenges in overcoming the complexity of the solid tumour microenvironment and the concerns that its long-term activity raises about potential unknown and unpredictable consequences in non-oncological diseases. RESULTS In the most recent studies, the senolytic potential of CAR T is becoming an exciting field of research. Still, experimental but promising results indeed indicate the clearance of senescent cells as an effective strategy to improve exercise capacity and metabolic dysfunction in physiological ageing, with long-term therapeutic and preventive effects. However, an effective expansion of a CAR T population requires a lympho-depleting chemotherapy prior to infusion. While this procedure sounds reasonable for rescue therapy of oncological diseases, it poses genotoxic risks that may not be justified for non-malignant diseases. Those represent the leading gaps for applying CAR T therapy in non-oncological diseases. CONCLUSION More is expected from current studies on the other classes of CAR cells now under investigation. Engineering NK cells and macrophages are candidates to improve cytotoxic and immunomodulating properties, potentially able to broaden application in solid tumours and non-oncological diseases. Finally, engineering autologous T cells in old individuals may generate biologically deteriorated CAR T clones with impaired function and unpredictable effects on cytokine release.
Collapse
Affiliation(s)
- Davide Ramoni
- Department of Internal Medicine, University of Genoa, Genoa, Italy
| | - Fabrizio Montecucco
- Department of Internal Medicine, University of Genoa, Genoa, Italy
- IRCCS Ospedale Policlinico San Martino Genoa, Italian Cardiovascular Network, Genoa, Italy
| | - Federico Carbone
- Department of Internal Medicine, University of Genoa, Genoa, Italy
- IRCCS Ospedale Policlinico San Martino Genoa, Italian Cardiovascular Network, Genoa, Italy
| |
Collapse
|
28
|
Nix MA, Wiita AP. Alternative target recognition elements for chimeric antigen receptor (CAR) T cells: beyond standard antibody fragments. Cytotherapy 2024; 26:729-738. [PMID: 38466264 DOI: 10.1016/j.jcyt.2024.02.024] [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: 10/16/2023] [Revised: 01/31/2024] [Accepted: 02/26/2024] [Indexed: 03/12/2024]
Abstract
BACKGROUND AIMS Chimeric antigen receptor T (CAR-T) cells are a remarkably efficacious, highly promising and rapidly evolving strategy in the field of immuno-oncology. The precision of these targeted cellular therapies is driven by the specificity of the antigen recognition element (the "binder") encoded in the CAR. This binder redirects these immune effector cells precisely toward a defined antigen on the surface of cancer cells, leading to T-cell receptor-independent tumor lysis. Currently, for tumor targeting most CAR-T cells are designed using single-chain variable fragments (scFvs) derived from murine or human immunoglobulins. However, there are several emerging alternative binder modalities that are finding increasing utility for improved CAR function beyond scFvs. METHODS Here we review the most recent developments in the use of non-canonical protein binding domains in CAR design, including nanobodies, DARPins, natural ligands, and de novo-designed protein elements. RESULTS Overall, we describe how new protein binder formats, with their unique structural properties and mechanisms of action, may possess key advantages over traditional scFv CAR designs. CONCLUSIONS These alternative binder designs may contribute to enhanced CAR-T therapeutic options and, ultimately, improved outcomes for cancer patients.
Collapse
Affiliation(s)
- Matthew A Nix
- Department of Laboratory Medicine, University of California, San Francisco, San Francisco, California, USA; Cartography Biosciences, South San Francisco, California, USA
| | - Arun P Wiita
- Department of Laboratory Medicine, University of California, San Francisco, San Francisco, California, USA; Department of Bioengineering and Therapeutic Sciences, University of California, San Francisco, San Francisco, California, USA; Chan Zuckerberg Biohub San Francisco, San Francisco, California, USA; Parker Institute for Cancer Immunotherapy, San Francisco, California, USA.
| |
Collapse
|
29
|
Kuznetsova AV, Glukhova XA, Popova OP, Beletsky IP, Ivanov AA. Contemporary Approaches to Immunotherapy of Solid Tumors. Cancers (Basel) 2024; 16:2270. [PMID: 38927974 PMCID: PMC11201544 DOI: 10.3390/cancers16122270] [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: 05/28/2024] [Revised: 06/11/2024] [Accepted: 06/15/2024] [Indexed: 06/28/2024] Open
Abstract
In recent years, the arrival of the immunotherapy industry has introduced the possibility of providing transformative, durable, and potentially curative outcomes for various forms of malignancies. However, further research has shown that there are a number of issues that significantly reduce the effectiveness of immunotherapy, especially in solid tumors. First of all, these problems are related to the protective mechanisms of the tumor and its microenvironment. Currently, major efforts are focused on overcoming protective mechanisms by using different adoptive cell therapy variants and modifications of genetically engineered constructs. In addition, a complex workforce is required to develop and implement these treatments. To overcome these significant challenges, innovative strategies and approaches are necessary to engineer more powerful variations of immunotherapy with improved antitumor activity and decreased toxicity. In this review, we discuss recent innovations in immunotherapy aimed at improving clinical efficacy in solid tumors, as well as strategies to overcome the limitations of various immunotherapies.
Collapse
Affiliation(s)
- Alla V. Kuznetsova
- Laboratory of Molecular and Cellular Pathology, Russian University of Medicine (Formerly A.I. Evdokimov Moscow State University of Medicine and Dentistry), Ministry of Health of the Russian Federation, Bld 4, Dolgorukovskaya Str, 1127006 Moscow, Russia; (A.V.K.); (O.P.P.)
- Koltzov Institute of Developmental Biology, Russian Academy of Sciences, 26 Vavilov Street, 119334 Moscow, Russia
| | - Xenia A. Glukhova
- Onni Biotechnologies Ltd., Aalto University Campus, Metallimiehenkuja 10, 02150 Espoo, Finland; (X.A.G.); (I.P.B.)
| | - Olga P. Popova
- Laboratory of Molecular and Cellular Pathology, Russian University of Medicine (Formerly A.I. Evdokimov Moscow State University of Medicine and Dentistry), Ministry of Health of the Russian Federation, Bld 4, Dolgorukovskaya Str, 1127006 Moscow, Russia; (A.V.K.); (O.P.P.)
| | - Igor P. Beletsky
- Onni Biotechnologies Ltd., Aalto University Campus, Metallimiehenkuja 10, 02150 Espoo, Finland; (X.A.G.); (I.P.B.)
| | - Alexey A. Ivanov
- Laboratory of Molecular and Cellular Pathology, Russian University of Medicine (Formerly A.I. Evdokimov Moscow State University of Medicine and Dentistry), Ministry of Health of the Russian Federation, Bld 4, Dolgorukovskaya Str, 1127006 Moscow, Russia; (A.V.K.); (O.P.P.)
| |
Collapse
|
30
|
D’Alò F, Bellesi S, Maiolo E, Alma E, Bellisario F, Malafronte R, Viscovo M, Campana F, Hohaus S. Novel Targets and Advanced Therapies in Diffuse Large B Cell Lymphomas. Cancers (Basel) 2024; 16:2243. [PMID: 38927948 PMCID: PMC11201587 DOI: 10.3390/cancers16122243] [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: 05/10/2024] [Revised: 06/07/2024] [Accepted: 06/14/2024] [Indexed: 06/28/2024] Open
Abstract
Since the introduction of rituximab in the late 1990s, significant progress has been made in advancing targeted therapies for B cell lymphomas, improving patients' chance of being cured and clinicians' therapeutic armamentarium. A better understanding of disease biology and pathogenic pathways, coupled with refinements in immunophenotypic and molecular diagnostics, have been instrumental in these achievements. While traditional chemotherapy remains fundamental in most cases, concerns surrounding chemorefractoriness and cumulative toxicities, particularly the depletion of the hemopoietic reserve, underscore the imperative for personalized treatment approaches. Integrating targeted agents, notably monoclonal antibodies, alongside chemotherapy has yielded heightened response rates and prolonged survival. A notable paradigm shift is underway with innovative-targeted therapies replacing cytotoxic drugs, challenging conventional salvage strategies like stem cell transplantation. This review examines the landscape of emerging targets for lymphoma cells and explores innovative therapies for diffuse large B cell lymphoma (DLBCL). From Chimeric Antigen Receptor-T cells to more potent monoclonal antibodies, antibody-drug conjugates, bispecific antibodies, checkpoint inhibitors, and small molecules targeting intracellular pathways, each modality offers promising avenues for therapeutic advancement. This review aims to furnish insights into their potential implications for the future of DLBCL treatment strategies.
Collapse
Affiliation(s)
- Francesco D’Alò
- Dipartimento di Scienze Radiologiche ed Ematologiche, Università Cattolica del Sacro Cuore, 00168 Rome, Italy; (F.B.); (R.M.); (M.V.); (F.C.); (S.H.)
- UOSD Malattie Linfoproliferative Extramidollari, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, 00168 Rome, Italy;
| | - Silvia Bellesi
- UOC Servizio e DH di Ematologia, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, 00168 Rome, Italy; (S.B.); (E.M.)
| | - Elena Maiolo
- UOC Servizio e DH di Ematologia, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, 00168 Rome, Italy; (S.B.); (E.M.)
| | - Eleonora Alma
- UOSD Malattie Linfoproliferative Extramidollari, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, 00168 Rome, Italy;
| | - Flaminia Bellisario
- Dipartimento di Scienze Radiologiche ed Ematologiche, Università Cattolica del Sacro Cuore, 00168 Rome, Italy; (F.B.); (R.M.); (M.V.); (F.C.); (S.H.)
- UOSD Malattie Linfoproliferative Extramidollari, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, 00168 Rome, Italy;
| | - Rosalia Malafronte
- Dipartimento di Scienze Radiologiche ed Ematologiche, Università Cattolica del Sacro Cuore, 00168 Rome, Italy; (F.B.); (R.M.); (M.V.); (F.C.); (S.H.)
- UOSD Malattie Linfoproliferative Extramidollari, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, 00168 Rome, Italy;
| | - Marcello Viscovo
- Dipartimento di Scienze Radiologiche ed Ematologiche, Università Cattolica del Sacro Cuore, 00168 Rome, Italy; (F.B.); (R.M.); (M.V.); (F.C.); (S.H.)
- UOSD Malattie Linfoproliferative Extramidollari, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, 00168 Rome, Italy;
| | - Fabrizia Campana
- Dipartimento di Scienze Radiologiche ed Ematologiche, Università Cattolica del Sacro Cuore, 00168 Rome, Italy; (F.B.); (R.M.); (M.V.); (F.C.); (S.H.)
- UOSD Malattie Linfoproliferative Extramidollari, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, 00168 Rome, Italy;
| | - Stefan Hohaus
- Dipartimento di Scienze Radiologiche ed Ematologiche, Università Cattolica del Sacro Cuore, 00168 Rome, Italy; (F.B.); (R.M.); (M.V.); (F.C.); (S.H.)
- UOSD Malattie Linfoproliferative Extramidollari, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, 00168 Rome, Italy;
| |
Collapse
|
31
|
Farhangnia P, Khorramdelazad H, Nickho H, Delbandi AA. Current and future immunotherapeutic approaches in pancreatic cancer treatment. J Hematol Oncol 2024; 17:40. [PMID: 38835055 PMCID: PMC11151541 DOI: 10.1186/s13045-024-01561-6] [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: 04/09/2024] [Accepted: 05/28/2024] [Indexed: 06/06/2024] Open
Abstract
Pancreatic cancer is a major cause of cancer-related death, but despondently, the outlook and prognosis for this resistant type of tumor have remained grim for a long time. Currently, it is extremely challenging to prevent or detect it early enough for effective treatment because patients rarely exhibit symptoms and there are no reliable indicators for detection. Most patients have advanced or spreading cancer that is difficult to treat, and treatments like chemotherapy and radiotherapy can only slightly prolong their life by a few months. Immunotherapy has revolutionized the treatment of pancreatic cancer, yet its effectiveness is limited by the tumor's immunosuppressive and hard-to-reach microenvironment. First, this article explains the immunosuppressive microenvironment of pancreatic cancer and highlights a wide range of immunotherapy options, including therapies involving oncolytic viruses, modified T cells (T-cell receptor [TCR]-engineered and chimeric antigen receptor [CAR] T-cell therapy), CAR natural killer cell therapy, cytokine-induced killer cells, immune checkpoint inhibitors, immunomodulators, cancer vaccines, and strategies targeting myeloid cells in the context of contemporary knowledge and future trends. Lastly, it discusses the main challenges ahead of pancreatic cancer immunotherapy.
Collapse
Affiliation(s)
- Pooya Farhangnia
- Reproductive Sciences and Technology Research Center, Department of Immunology, School of Medicine, Iran University of Medical Sciences, Tehran, Iran
- Immunology Research Center, Institute of Immunology and Infectious Diseases, Iran University of Medical Sciences, Tehran, Iran
- Department of Immunology, School of Medicine, Iran University of Medical Sciences, Tehran, Iran
- Immunology Board for Transplantation and Cell-Based Therapeutics (ImmunoTACT), Universal Scientific Education and Research Network (USERN), Tehran, Iran
| | - Hossein Khorramdelazad
- Department of Immunology, School of Medicine, Rafsanjan University of Medical Sciences, Rafsanjan, Iran
| | - Hamid Nickho
- Immunology Research Center, Institute of Immunology and Infectious Diseases, Iran University of Medical Sciences, Tehran, Iran
- Department of Immunology, School of Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Ali-Akbar Delbandi
- Reproductive Sciences and Technology Research Center, Department of Immunology, School of Medicine, Iran University of Medical Sciences, Tehran, Iran.
- Immunology Research Center, Institute of Immunology and Infectious Diseases, Iran University of Medical Sciences, Tehran, Iran.
- Department of Immunology, School of Medicine, Iran University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
32
|
Sebastian G. How I treat newly diagnosed acute lymphoblastic leukemia. Clin Hematol Int 2024; 6:51-61. [PMID: 38817308 PMCID: PMC11088446 DOI: 10.46989/001c.117026] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2023] [Accepted: 05/31/2023] [Indexed: 06/01/2024] Open
Abstract
Treatment algorithms differ for adult patients with Philadelphia-negative (Ph-) and Philadelphia-positive (Ph+) acute lymphoblastic leukemia (ALL). For Ph- ALL intensive induction-consolidation chemotherapy using "pediatric-inspired" protocols is a standard of care. Allogeneic hematopoietic cell transplantation (allo-HCT) from either an HLA-matched sibling, unrelated or haploidentical donor should be considered for patients with high estimated risk of relapse. Inadequate response at the level of measurable residual disease (MRD) is the strongest adverse prognostic factor. Patients with B-ALL and detectable MRD should be treated with blinatumomab. In the future, the use of blinatumomab and/or inotuzumab ozogamycin in addition to first-line chemotherapy may become a new standard of care reducing the role of allo-HCT. For patients with Ph+ ALL, tyrosine kinase inhibitors (TKI) are the most important components of treatment protocols, while the intensity of chemotherapy may be reduced. Allo-HCT is recommended for all patients treated with imatinib along with low-intensity chemotherapy. Results of phase-II studies using front-line dasatinib or ponatinib in sequence or in combination with blinatumomab are very promising. Such a strategy may allow the avoidance of systemic chemotherapy. The future role of allo-HCT in this context appears uncertain.
Collapse
Affiliation(s)
- Giebel Sebastian
- Bone Marrow Transplantation and Onco-HematologyMaria Sklodowska-Curie National Research Institute of Oncology Gliwice Branch
| |
Collapse
|
33
|
Hassan SH, Alshahrani MY, Saleh RO, Mohammed BA, Kumar A, Almalki SG, Alkhafaji AT, Ghildiyal P, Al-Tameemi AR, Elawady A. A new vision of the efficacy of both CAR-NK and CAR-T cells in treating cancers and autoimmune diseases. Med Oncol 2024; 41:127. [PMID: 38656354 DOI: 10.1007/s12032-024-02362-0] [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: 02/21/2024] [Accepted: 03/19/2024] [Indexed: 04/26/2024]
Abstract
Chimeric Antigen Receptor (CAR) based therapies are becoming increasingly important in treating patients. CAR-T cells have been shown to be highly effective in the treatment of hematological malignancies. However, harmful therapeutic barriers have been identified, such as the potential for graft-versus-host disease (GVHD), neurotoxicity, and cytokine release syndrome (CRS). As a result, CAR NK-cell therapy is expected to be a new therapeutic option. NK cells act as cytotoxic lymphocytes, supporting the innate immune response against autoimmune diseases and cancer cells by precisely detecting and eliminating malignant cells. Genetic modification of these cells provides a dual approach to the treatment of AD and cancer. It can be used through both CAR-independent and CAR-dependent mechanisms. The use of CAR-based cell therapies has been successful in treating cancer patients, leading to further investigation of this innovative treatment for alternative diseases, including AD. The complementary roles of CAR T and CAR NK cells have stimulated exploration in this area. Our study examines the latest research on the therapeutic effectiveness of these cells in treating both cancer and ADs.
Collapse
Affiliation(s)
- Salim Hussein Hassan
- Community Health Department, Technical Institute of Karbala, AL-Furat Al-Awsat Technical University, Najaf, Iraq.
| | - Mohammad Y Alshahrani
- Department of Clinical Laboratory Sciences, College of Applied Medical Science, King Khalid University, Abha, Saudi Arabia
| | - Raed Obaid Saleh
- Department of Medical Laboratory Techniques, Al-Maarif University College, Al-Anbar, Iraq
| | | | - Abhinav Kumar
- Department of Nuclear and Renewable Energy, Ural Federal University Named After the First President of Russia Boris Yeltsin, Ekaterinburg, 620002, Russia
| | - Sami G Almalki
- Department of Medical Laboratory Sciences, College of Applied Medical Sciences, Majmaah University, 11952, Majmaah, Saudi Arabia
| | | | - Pallavi Ghildiyal
- Uttaranchal Institute of Pharmaceutical Sciences, Uttaranchal University, Dehradun, India
| | | | - Ahmed Elawady
- College of Technical Engineering, The Islamic University, Najaf, Iraq
- College of Technical Engineering, The Islamic University of Al Diwaniyah, Al Diwaniyah, Iraq
- College of Technical Engineering, The Islamic University of Babylon, Babylon, Iraq
| |
Collapse
|
34
|
Guijarro-Albaladejo B, Marrero-Cepeda C, Rodríguez-Arbolí E, Sierro-Martínez B, Pérez-Simón JA, García-Guerrero E. Chimeric antigen receptor (CAR) modified T Cells in acute myeloid leukemia: limitations and expectations. Front Cell Dev Biol 2024; 12:1376554. [PMID: 38694825 PMCID: PMC11061469 DOI: 10.3389/fcell.2024.1376554] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2024] [Accepted: 04/04/2024] [Indexed: 05/04/2024] Open
Abstract
Acute myeloid leukemia (AML) is an aggressive hematologic malignancy with a poor prognosis despite the advent of novel therapies. Consequently, a major need exists for new therapeutic options, particularly for patients with relapsed/refractory (R/R) AML. In recent years, it has been possible to individualize the treatment of a subgroup of patients, particularly with the emergence of multiple targeted therapies. Nonetheless, a considerable number of patients remain without therapeutic options, and overall prognosis remains poor because of a high rate of disease relapse. In this sense, cellular therapies, especially chimeric antigen receptor (CAR)-T cell therapy, have dramatically shifted the therapeutic options for other hematologic malignancies, such as diffuse large B cell lymphoma and acute lymphoblastic leukemia. In contrast, effectively treating AML with CAR-based immunotherapy poses major biological and clinical challenges, most of them derived from the unmet need to identify target antigens with expression restricted to the AML blast without compromising the viability of the normal hematopoietic stem cell counterpart. Although those limitations have hampered CAR-T cell therapy translation to the clinic, there are several clinical trials where target antigens, such as CD123, CLL-1 or CD33 are being used to treat AML patients showing promising results. Moreover, there are continuing efforts to enhance the specificity and efficacy of CAR-T cell therapy in AML. These endeavors encompass the exploration of novel avenues, including the development of dual CAR-T cells and next-generation CAR-T cells, as well as the utilization of gene editing tools to mitigate off-tumor toxicities. In this review, we will summarize the ongoing clinical studies and the early clinical results reported with CAR-T cells in AML, as well as highlight CAR-T cell limitations and the most recent approaches to overcome these barriers. We will also discuss how and when CAR-T cells should be used in the context of AML.
Collapse
Affiliation(s)
- Beatriz Guijarro-Albaladejo
- Instituto de Biomedicina de Sevilla, IBiS/Hospital Universitario Virgen del Rocío/CSIC/Universidad de Sevilla, Servicio de Hematología, Hospital Universitario Virgen del Rocío, Seville, Spain
| | - Cristina Marrero-Cepeda
- Unidad de Gestión Clínica de Hematología, Instituto de Biomedicina de Sevilla, IBiS/Hospital Universitario Virgen del Rocío/CSIC/Universidad de Sevilla, Seville, Spain
| | - Eduardo Rodríguez-Arbolí
- Unidad de Gestión Clínica de Hematología, Instituto de Biomedicina de Sevilla, IBiS/Hospital Universitario Virgen del Rocío/CSIC/Universidad de Sevilla, Seville, Spain
| | - Belén Sierro-Martínez
- Instituto de Biomedicina de Sevilla, IBiS/Hospital Universitario Virgen del Rocío/CSIC/Universidad de Sevilla, Servicio de Hematología, Hospital Universitario Virgen del Rocío, Seville, Spain
| | - José Antonio Pérez-Simón
- Unidad de Gestión Clínica de Hematología, Instituto de Biomedicina de Sevilla, IBiS/Hospital Universitario Virgen del Rocío/CSIC/Universidad de Sevilla, Seville, Spain
| | - Estefanía García-Guerrero
- Instituto de Biomedicina de Sevilla, IBiS/Hospital Universitario Virgen del Rocío/CSIC/Universidad de Sevilla, Servicio de Hematología, Hospital Universitario Virgen del Rocío, Seville, Spain
| |
Collapse
|
35
|
Rotte A. Editorial: Emerging learnings in cell therapy: novel binding domains, universal CAR-T cells, and more. Front Oncol 2024; 14:1404376. [PMID: 38686191 PMCID: PMC11056576 DOI: 10.3389/fonc.2024.1404376] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2024] [Accepted: 03/29/2024] [Indexed: 05/02/2024] Open
Affiliation(s)
- Anand Rotte
- Department of Clinical and Regulatory Affairs, Arcellx Inc, Redwood City, CA, United States
| |
Collapse
|
36
|
Pu Y, Zhao Y, Qi Y, Liu Y, Zhang M, Xiao X, Lyu H, Meng J, Zhu H, Xu K, Han W, Zhao M. Multi-centers experience using therapeutic plasma exchange for corticosteroid/tocilizumab-refractory cytokine release syndrome following CAR-T therapy. Int Immunopharmacol 2024; 130:111761. [PMID: 38422769 DOI: 10.1016/j.intimp.2024.111761] [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: 01/08/2024] [Revised: 02/14/2024] [Accepted: 02/23/2024] [Indexed: 03/02/2024]
Abstract
The chimeric antigen receptor T (CAR-T) cell therapy significantly enhances the prognosis of various hematologic malignancies; however, the systemic expansion of CAR-T cells also gives rise to severe cytokine release syndrome (CRS), and immune effector cell-associated neurotoxicity syndrome (ICANS). Despite the successful application of corticosteroids and tocilizumab in alleviating severe CRS in most patients, there are still individuals who experience life-threatening CRS without responding to the aforementioned therapies. In our retrospective cohort, we conducted an analysis of clinical and laboratory parameters, including inflammatory cytokines, in 17 patients from three centers who underwent therapeutic plasma exchange (TPE) for refractory CRS with or without ICANS following CAR-T products treatment. Our findings demonstrate a significant improvement in both clinical symptoms and laboratory parameters subsequent to TPE treatment. The rapid decrease in temperature and levels of inflammatory indexes indicates the remarkable scavenging efficacy of TPE against cytokine storm following CAR-T therapy. In conclusion, TPE may serve as a valuable and safe adjunct to corticosteroids and tocilizumab in the management of severe CRS resulting from CAR-T cell infusion. We eagerly await further prospective studies to validate this finding.
Collapse
Affiliation(s)
- Yedi Pu
- Department of Hematology, Tianjin First Central Hospital, Tianjin 300380, China
| | - Yifan Zhao
- The First Central Clinical College of Tianjin Medical University, Tianjin 300380, China
| | - Yuekun Qi
- Department of Hematology, The Affiliated Hospital of Xuzhou Medical University, Xuzhou 221002, China
| | - Yang Liu
- Department of Bio-therapeutic, The First Medical Center, Chinese PLA General Hospital, Beijing 100853, China
| | - Meng Zhang
- Department of Hematology, Tianjin First Central Hospital, Tianjin 300380, China
| | - Xia Xiao
- Department of Hematology, Tianjin First Central Hospital, Tianjin 300380, China
| | - Hairong Lyu
- Department of Hematology, Tianjin First Central Hospital, Tianjin 300380, China
| | - Juanxia Meng
- Department of Hematology, Tianjin First Central Hospital, Tianjin 300380, China
| | - Haibo Zhu
- Department of Hematology, Tianjin First Central Hospital, Tianjin 300380, China
| | - Kailin Xu
- Department of Hematology, The Affiliated Hospital of Xuzhou Medical University, Xuzhou 221002, China.
| | - Weidong Han
- Department of Bio-therapeutic, The First Medical Center, Chinese PLA General Hospital, Beijing 100853, China.
| | - Mingfeng Zhao
- Department of Hematology, Tianjin First Central Hospital, Tianjin 300380, China.
| |
Collapse
|
37
|
Ho M, Zanwar S, Paludo J. Chimeric antigen receptor T-cell therapy in hematologic malignancies: Successes, challenges, and opportunities. Eur J Haematol 2024; 112:197-210. [PMID: 37545132 DOI: 10.1111/ejh.14074] [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: 06/01/2023] [Revised: 07/25/2023] [Accepted: 07/25/2023] [Indexed: 08/08/2023]
Abstract
The success of chimeric antigen receptor T-cell (CAR-T) therapy in hematologic malignancies has realized a longstanding effort toward harnessing the immune system to fight cancer in a truly personalized fashion. Second generation chimeric antigen receptors (CAR) incorporating co-stimulatory molecules like 4-1BB or CD28 were able to overcome some of the hindrances with initial CAR constructs resulting in efficacious products. Many second-generation CAR-T products have been approved in the treatment of relapsed/refractory hematologic malignancies including multiple myeloma (MM), non-Hodgkin lymphoma (NHL), and acute lymphoblastic leukemia. However, challenges remain in optimizing the manufacturing, timely access, limiting the toxicity from CAR-T infusions and improving sustainability of responses derived with CAR-T therapy. Here, we summarize the clinical trial data leading to approval CAR-T therapies in MM and NHL, discuss the limitations with current CAR-T therapy strategies and review emerging strategies for overcoming these limitations.
Collapse
Affiliation(s)
- Matthew Ho
- Division of General Internal Medicine, Department of Medicine, Mayo Clinic, Rochester, Minnesota, USA
| | - Saurabh Zanwar
- Division of Hematology, Department of Medicine, Mayo Clinic, Rochester, Minnesota, USA
| | - Jonas Paludo
- Division of General Internal Medicine, Department of Medicine, Mayo Clinic, Rochester, Minnesota, USA
| |
Collapse
|
38
|
Pievani A, Biondi M, Tettamanti S, Biondi A, Dotti G, Serafini M. CARs are sharpening their weapons. J Immunother Cancer 2024; 12:e008275. [PMID: 38296592 PMCID: PMC10831441 DOI: 10.1136/jitc-2023-008275] [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] [Accepted: 01/07/2024] [Indexed: 02/03/2024] Open
Abstract
Abstract
Collapse
Affiliation(s)
- Alice Pievani
- Tettamanti Center and Pediatrics, Fondazione IRCCS San Gerardo dei Tintori, Monza, Italy
| | - Marta Biondi
- Tettamanti Center and Pediatrics, Fondazione IRCCS San Gerardo dei Tintori, Monza, Italy
- School of Medicine and Surgery, University of Milan-Bicocca, Milano, Italy
| | - Sarah Tettamanti
- Tettamanti Center and Pediatrics, Fondazione IRCCS San Gerardo dei Tintori, Monza, Italy
| | - Andrea Biondi
- Tettamanti Center and Pediatrics, Fondazione IRCCS San Gerardo dei Tintori, Monza, Italy
- School of Medicine and Surgery, University of Milan-Bicocca, Milano, Italy
| | - Gianpietro Dotti
- Lineberger Comprehensive Cancer Center, University of North Carolina, Chapel Hill, North Carolina, USA
| | - Marta Serafini
- Tettamanti Center and Pediatrics, Fondazione IRCCS San Gerardo dei Tintori, Monza, Italy
- School of Medicine and Surgery, University of Milan-Bicocca, Milano, Italy
| |
Collapse
|
39
|
Tawfeeq C, Song J, Khaniya U, Madej T, Wang J, Youkharibache P, Abrol R. Towards a structural and functional analysis of the immunoglobulin-fold proteome. ADVANCES IN PROTEIN CHEMISTRY AND STRUCTURAL BIOLOGY 2024; 138:135-178. [PMID: 38220423 DOI: 10.1016/bs.apcsb.2023.11.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/16/2024]
Abstract
The immunoglobulin fold (Ig fold) domain is a super-secondary structural motif consisting of a sandwich with two layers of β-sheets that is present in many proteins with very diverse biological functions covering a wide range of physiological processes. This domain presents a modular architecture built with β strands connected by variable length loops that has a highly conserved structural core of four β-strands and quite variable β-sheet extensions in the two sandwich layers that enable both divergent and convergent evolutionary mechanisms in the known Ig fold proteome. The central role of this Ig fold's structural plasticity in the evolutionary success of antibodies in our immune system is well established. Nature has also utilized this Ig fold in all domains of life in many different physiological contexts that go way beyond the immune system. Here we will present a structural and functional overview of the utilization of the Ig fold in different biological processes and in different cellular contexts to highlight some of the innumerable ways that this structural motif can interact in multidomain proteins to enable their diversity of functions. This includes shareable specific protein structure visualizations behind those functions that serve as starting points for further explorations of the biomolecular interactions spanning the Ig fold proteome. This overview also highlights how this Ig fold is being utilized through natural adaptation, engineering, and even building from scratch for a range of biotechnological applications.
Collapse
Affiliation(s)
- Caesar Tawfeeq
- Department of Chemistry and Biochemistry, California State University Northridge, Northridge, United States
| | - James Song
- National Center for Biotechnology Information, National Library of Medicine, National Institutes of Health, Bethesda, United States
| | - Umesh Khaniya
- Cancer Data Science Laboratory, National Cancer Institute, National Institutes of Health, Bethesda, United States
| | - Thomas Madej
- National Center for Biotechnology Information, National Library of Medicine, National Institutes of Health, Bethesda, United States
| | - Jiyao Wang
- National Center for Biotechnology Information, National Library of Medicine, National Institutes of Health, Bethesda, United States
| | - Philippe Youkharibache
- Cancer Data Science Laboratory, National Cancer Institute, National Institutes of Health, Bethesda, United States.
| | - Ravinder Abrol
- Department of Chemistry and Biochemistry, California State University Northridge, Northridge, United States.
| |
Collapse
|
40
|
Bardhan M, Dey D, Suresh V, Javed B, Venur VA, Joe N, Kalidindi R, Ozair A, Khan M, Mahtani R, Lo S, Odia Y, Ahluwalia MS. An overview of the therapeutic strategies for neoplastic meningitis due to breast cancer: when and why? Expert Rev Neurother 2024; 24:77-103. [PMID: 38145503 DOI: 10.1080/14737175.2023.2293223] [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: 10/04/2023] [Accepted: 12/06/2023] [Indexed: 12/27/2023]
Abstract
INTRODUCTION Neoplastic meningitis (NM), also known as leptomeningeal carcinomatosis, is characterized by the infiltration of tumor cells into the meninges, and poses a significant therapeutic challenge owing to its aggressive nature and limited treatment options. Breast cancer is a common cause of NM among solid tumors, further highlighting the urgent need to explore effective therapeutic strategies. This review aims to provide insights into the evolving landscape of NM therapy in breast cancer by collating existing research, evaluating current treatments, and identifying potential emerging therapeutic options. AREAS COVERED This review explores the clinical features, therapeutic strategies, recent advances, and challenges of managing NM in patients with breast cancer. Its management includes multimodal strategies, including systemic and intrathecal chemotherapy, radiation therapy, and supportive care. This review also emphasizes targeted drug options and optimal drug concentrations, and discusses emerging therapies. Additionally, it highlights the variability in treatment outcomes and the potential of combination regimens to effectively manage NM in breast cancer. EXPERT OPINION Challenges in treating NM include debates over clinical trial end points and the management of adverse effects. Drug resistance and low response rates are significant hurdles, particularly inHER2-negative breast cancer. The development of more precise and cost-effective medications with improved selectivity is crucial. Additionally, global efforts are needed for infrastructure development and cancer control considering the diverse nature of the disease.
Collapse
Affiliation(s)
- Mainak Bardhan
- Department of Medical Oncology, Miami Cancer Institute, Baptist Health South Florida, Miami, FL, USA
| | | | - Vinay Suresh
- King George's Medical University, Lucknow, Uttar Pradesh, India
| | - Binish Javed
- Atal Bihari Vajpayee Institute of Medical Sciences and Dr. Ram Manohar Lohia Hospital, New Delhi, India
| | - Vyshak Alva Venur
- Seattle Cancer Care Alliance, Fred Hutchinson Cancer Center, University of Washington, Seattle, WA, USA
| | - Neha Joe
- St John's Medical College Hospital, Bengaluru, India
| | | | - Ahmad Ozair
- Department of Medical Oncology, Miami Cancer Institute, Baptist Health South Florida, Miami, FL, USA
- Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, USA
| | | | - Reshma Mahtani
- Department of Medical Oncology, Miami Cancer Institute, Baptist Health South Florida, Miami, FL, USA
| | - Simon Lo
- Department of Radiation Oncology, University of Washington School of Medicine, Seattle, WA, USA
| | - Yazmin Odia
- Department of Medical Oncology, Miami Cancer Institute, Baptist Health South Florida, Miami, FL, USA
| | - Manmeet S Ahluwalia
- Department of Medical Oncology, Miami Cancer Institute, Baptist Health South Florida, Miami, FL, USA
- Herbert Wertheim College of Medicine, Florida International University, Miami, FL, USA
| |
Collapse
|
41
|
Ding H, Wu Y. CAR-T Therapy in Relapsed Refractory Multiple Myeloma. Curr Med Chem 2024; 31:4362-4382. [PMID: 37779413 PMCID: PMC11340289 DOI: 10.2174/0109298673268932230920063933] [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: 06/25/2023] [Revised: 08/06/2023] [Accepted: 08/18/2023] [Indexed: 10/03/2023]
Abstract
Multiple myeloma is a plasma cell neoplasm. The emergence of proteasome inhibitors, immunomodulatory drugs, and anti-CD38 monoclonal antibodies has improved the prognosis of multiple myeloma patients. However, some patients are still insensitive to conventional therapy or frequently relapse after remission. Chemotherapy based on proteasome inhibitors or immunomodulatory drugs is ineffective in controlling the progression of relapsed refractory multiple myeloma. No consensus has been reached on treating relapsed refractory multiple myeloma to date. Recently chimeric antigen receptor T cells therapy has shown promising results that could achieve rapid remissions of patients and improve their prognoses. Additionally, most patients in chimeric antigen receptor T cell clinical trials were triple-refractory multiple myeloma patients, indicating that chimeric antigen receptor T cell immunotherapy could overcome drug resistance to new drugs. Since single immunotherapies are prone to acquired resistance, combination immunotherapies based on emerging immunotherapies may solve this issue. Achieving complete remission and minimal residual disease negative status as soon as possible is beneficial to patients. This paper reviewed the main chimeric antigen receptor T cell products in relapsed refractory multiple myeloma, and it explained the drug resistance mechanism and improvement methods of chimeric antigen receptor T cells therapy. This review summarized the best beneficiaries of chimeric antigen receptor T cell therapy and the salvage treatment of disease recurrence after chimeric antigen receptor T cell therapy, providing some ideas for the clinical application of chimeric antigen receptor T cells.
Collapse
Affiliation(s)
- Hong Ding
- Department of Hematology, West China Hospital, Sichuan University, China
| | - Yu Wu
- Department of Hematology, West China Hospital, Sichuan University, China
| |
Collapse
|
42
|
Goutnik M, Iakovidis A, Still MEH, Moor RSF, Melnick K, Yan S, Abbas M, Huang J, Ghiaseddin AP. Advancements in chimeric antigen receptor-expressing T-cell therapy for glioblastoma multiforme: Literature review and future directions. Neurooncol Adv 2024; 6:vdae025. [PMID: 38486856 PMCID: PMC10939440 DOI: 10.1093/noajnl/vdae025] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/17/2024] Open
Abstract
Glioblastoma multiforme (GBM) is an aggressive cancer that has been difficult to treat and often requires multimodal therapy consisting of surgery, radiotherapy, and chemotherapy. Chimeric antigen receptor-expressing (CAR-T) cells have been efficacious in treating hematological malignancies, resulting in several FDA-approved therapies. CAR-T cells have been more recently studied for the treatment of GBM, with some promising preclinical and clinical results. The purpose of this literature review is to highlight the commonly targeted antigens, results of clinical trials, novel modifications, and potential solutions for challenges that exist for CAR-T cells to become more widely implemented and effective in eradicating GBM.
Collapse
Affiliation(s)
- Michael Goutnik
- Department of Neurosurgery, College of Medicine, University of Florida, Gainesville, Florida, USA
| | - Alexandria Iakovidis
- Department of Neurosurgery, College of Medicine, University of Florida, Gainesville, Florida, USA
| | - Megan E H Still
- Department of Neurosurgery, College of Medicine, University of Florida, Gainesville, Florida, USA
| | - Rachel S F Moor
- Department of Neurosurgery, College of Medicine, University of Florida, Gainesville, Florida, USA
| | - Kaitlyn Melnick
- Department of Neurosurgery, College of Medicine, University of Florida, Gainesville, Florida, USA
| | - Sandra Yan
- Department of Neurosurgery, College of Medicine, University of Florida, Gainesville, Florida, USA
| | - Muhammad Abbas
- Department of Neurosurgery, College of Medicine, University of Florida, Gainesville, Florida, USA
| | - Jianping Huang
- Department of Neurosurgery, College of Medicine, University of Florida, Gainesville, Florida, USA
| | - Ashley P Ghiaseddin
- Department of Neurosurgery, College of Medicine, University of Florida, Gainesville, Florida, USA
| |
Collapse
|
43
|
Chamorro DF, Somes LK, Hoyos V. Engineered Adoptive T-Cell Therapies for Breast Cancer: Current Progress, Challenges, and Potential. Cancers (Basel) 2023; 16:124. [PMID: 38201551 PMCID: PMC10778447 DOI: 10.3390/cancers16010124] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2023] [Revised: 12/19/2023] [Accepted: 12/21/2023] [Indexed: 01/12/2024] Open
Abstract
Breast cancer remains a significant health challenge, and novel treatment approaches are critically needed. This review presents an in-depth analysis of engineered adoptive T-cell therapies (E-ACTs), an innovative frontier in cancer immunotherapy, focusing on their application in breast cancer. We explore the evolving landscape of chimeric antigen receptor (CAR) and T-cell receptor (TCR) T-cell therapies, highlighting their potential and challenges in targeting breast cancer. The review addresses key obstacles such as target antigen selection, the complex breast cancer tumor microenvironment, and the persistence of engineered T-cells. We discuss the advances in overcoming these barriers, including strategies to enhance T-cell efficacy. Finally, our comprehensive analysis of the current clinical trials in this area provides insights into the future possibilities and directions of E-ACTs in breast cancer treatment.
Collapse
Affiliation(s)
- Diego F. Chamorro
- Center for Cell and Gene Therapy, Baylor College of Medicine, Houston, TX 77030, USA; (D.F.C.); (L.K.S.)
| | - Lauren K. Somes
- Center for Cell and Gene Therapy, Baylor College of Medicine, Houston, TX 77030, USA; (D.F.C.); (L.K.S.)
| | - Valentina Hoyos
- Center for Cell and Gene Therapy, Baylor College of Medicine, Houston, TX 77030, USA; (D.F.C.); (L.K.S.)
- Dan L. Duncan Comprehensive Cancer Center, Baylor College of Medicine, Houston, TX 77030, USA
| |
Collapse
|
44
|
Vianzon VV, Hanson RM, Garg I, Joseph GJ, Rogers LM. Rank aggregation of independent genetic screen results highlights new strategies for adoptive cellular transfer therapy of cancer. Front Immunol 2023; 14:1235131. [PMID: 38143765 PMCID: PMC10748423 DOI: 10.3389/fimmu.2023.1235131] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2023] [Accepted: 11/20/2023] [Indexed: 12/26/2023] Open
Abstract
Efficient intratumoral infiltration of adoptively transferred cells is a significant barrier to effectively treating solid tumors with adoptive cellular transfer (ACT) therapies. Our recent forward genetic, whole-genome screen identified T cell-intrinsic gene candidates that may improve tumor infiltration of T cells. Here, results are combined with five independent genetic screens using rank aggregation to improve rigor. This resulted in a combined total of 1,523 candidate genes - including 1,464 genes not currently being evaluated as therapeutic targets - that may improve tumor infiltration of T cells. Gene set enrichment analysis of a published human dataset shows that these gene candidates are differentially expressed in tumor infiltrating compared to circulating T cells, supporting translational potential. Importantly, adoptive transfer of T cells overexpressing gain-of-function candidates (AAK1ΔN125, SPRR1B, and EHHADH) into tumor-bearing mice resulted in increased T cell infiltration into tumors. These novel gene candidates may be considered as potential therapeutic candidates that can aid adoptive cellular therapy in improving T cell infiltration into solid tumors.
Collapse
Affiliation(s)
| | | | | | | | - Laura M. Rogers
- Department of Immunology, Mayo Clinic, Rochester, MN, United States
| |
Collapse
|
45
|
Pessach I, Nagler A. Leukapheresis for CAR-T cell production and therapy. Transfus Apher Sci 2023; 62:103828. [PMID: 37838564 DOI: 10.1016/j.transci.2023.103828] [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] [Indexed: 10/16/2023]
Abstract
Chimeric antigen receptor (CAR) T-cell therapy is an effective, individualized immunotherapy, and novel treatment for hematologic malignancies. Six commercial CAR-T cell products are currently approved for lymphatic malignancies and multiple myeloma. In addition, an increasing number of clinical centres produce CAR-T cells on-site, which enable the administration of CAR-T cells on site. The CAR-T cell products are either fresh or cryopreserved. Manufacturing CAR-T cells is a complicated process that begins with leukapheresis to obtain T cells from the patient's peripheral blood. An optimal leukapheresis product is crucial step for a successful CAR-T cell therapy; therefore, it is imperative to understand the factors that may affect the quality or T cells. The leukapheresis for CAR-T cell production is well tolerated and safe for both paediatric and adult patients and CAR-Τ cell therapy presents high clinical response rate in many studies. CAR-T cell therapy is under continuous improvement, and it has transformed into an almost standard procedure in clinical haematology and stem cell transplantation facilities that provide both autologous and allogeneic stem cell transplantations. In patients suffering from advanced haematological malignancies, CAR-T cell therapy shows incredible antitumor efficacy. Even after a single infusion of autologous CD19-targeting CAR-T cells in patients with relapsed or refractory diffuse large B cell lymphoma (DLBCL) and acute lymphoblastic leukaemia (ALL), long lasting remission is observed, and a fraction of the patients are being cured. Future novel constructs are being developed with better T cell persistence and better expansion. New next-generation CAR-T cells are currently designed to avoid toxicities such as cytokine release syndrome and neurotoxicity.
Collapse
Affiliation(s)
- Ilias Pessach
- Hematology Department, Athens Medical Center, Athens, Greece
| | - Arnon Nagler
- Hematology Division, Chaim Sheba Medical Center, Israel.
| |
Collapse
|
46
|
Miliotou AN, Georgiou-Siafis SK, Ntenti C, Pappas IS, Papadopoulou LC. Recruiting In Vitro Transcribed mRNA against Cancer Immunotherapy: A Contemporary Appraisal of the Current Landscape. Curr Issues Mol Biol 2023; 45:9181-9214. [PMID: 37998753 PMCID: PMC10670245 DOI: 10.3390/cimb45110576] [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: 10/15/2023] [Revised: 11/05/2023] [Accepted: 11/14/2023] [Indexed: 11/25/2023] Open
Abstract
Over 100 innovative in vitro transcribed (IVT)-mRNAs are presently undergoing clinical trials, with a projected substantial impact on the pharmaceutical market in the near future. Τhe idea behind this is that after the successful cellular internalization of IVT-mRNAs, they are subsequently translated into proteins with therapeutic or prophylactic relevance. Simultaneously, cancer immunotherapy employs diverse strategies to mobilize the immune system in the battle against cancer. Therefore, in this review, the fundamental principles of IVT-mRNA to its recruitment in cancer immunotherapy, are discussed and analyzed. More specifically, this review paper focuses on the development of mRNA vaccines, the exploitation of neoantigens, as well as Chimeric Antigen Receptor (CAR) T-Cells, showcasing their clinical applications and the ongoing trials for the development of next-generation immunotherapeutics. Furthermore, this study investigates the synergistic potential of combining the CAR immunotherapy and the IVT-mRNAs by introducing our research group novel, patented delivery method that utilizes the Protein Transduction Domain (PTD) technology to transduce the IVT-mRNAs encoding the CAR of interest into the Natural Killer (NK)-92 cells, highlighting the potential for enhancing the CAR NK cell potency, efficiency, and bioenergetics. While IVT-mRNA technology brings exciting progress to cancer immunotherapy, several challenges and limitations must be acknowledged, such as safety, toxicity, and delivery issues. This comprehensive exploration of IVT-mRNA technology, in line with its applications in cancer therapeutics, offers valuable insights into the opportunities and challenges in the evolving landscape of cancer immunotherapy, setting the stage for future advancements in the field.
Collapse
Affiliation(s)
- Androulla N. Miliotou
- Laboratory of Pharmacology, School of Pharmacy, Faculty of Health Sciences, Aristotle University of Thessaloniki, 54124 Thessaloniki, Macedonia, Greece; (A.N.M.); (S.K.G.-S.); (C.N.)
- Department of Health Sciences, KES College, 1055 Nicosia, Cyprus
- Faculty of Pharmacy, Department of Health Sciences, University of Nicosia, 1700 Nicosia, Cyprus
| | - Sofia K. Georgiou-Siafis
- Laboratory of Pharmacology, School of Pharmacy, Faculty of Health Sciences, Aristotle University of Thessaloniki, 54124 Thessaloniki, Macedonia, Greece; (A.N.M.); (S.K.G.-S.); (C.N.)
- Laboratory of Pharmacology and Toxicology, Faculty of Veterinary Medicine, University of Thessaly, 43100 Karditsa, Thessaly, Greece;
| | - Charikleia Ntenti
- Laboratory of Pharmacology, School of Pharmacy, Faculty of Health Sciences, Aristotle University of Thessaloniki, 54124 Thessaloniki, Macedonia, Greece; (A.N.M.); (S.K.G.-S.); (C.N.)
- 1st Laboratory of Pharmacology, School of Medicine, Faculty of Health Sciences, Aristotle University of Thessaloniki, 54124 Thessaloniki, Macedonia, Greece
| | - Ioannis S. Pappas
- Laboratory of Pharmacology and Toxicology, Faculty of Veterinary Medicine, University of Thessaly, 43100 Karditsa, Thessaly, Greece;
| | - Lefkothea C. Papadopoulou
- Laboratory of Pharmacology, School of Pharmacy, Faculty of Health Sciences, Aristotle University of Thessaloniki, 54124 Thessaloniki, Macedonia, Greece; (A.N.M.); (S.K.G.-S.); (C.N.)
| |
Collapse
|
47
|
Garg S, Ni W, Griffin JD, Sattler M. Chimeric Antigen Receptor T Cell Therapy in Acute Myeloid Leukemia: Trials and Tribulations. Hematol Rep 2023; 15:608-626. [PMID: 37987319 PMCID: PMC10660693 DOI: 10.3390/hematolrep15040063] [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: 07/18/2023] [Revised: 08/01/2023] [Accepted: 11/08/2023] [Indexed: 11/22/2023] Open
Abstract
Acute myeloid leukemia (AML) is a heterogeneous hematological malignancy that is often associated with relapse and drug resistance after standard chemotherapy or targeted therapy, particularly in older patients. Hematopoietic stem cell transplants are looked upon as the ultimate salvage option with curative intent. Adoptive cell therapy using chimeric antigen receptors (CAR) has shown promise in B cell malignancies and is now being investigated in AML. Initial clinical trials have been disappointing in AML, and we review current strategies to improve efficacy for CAR approaches. The extensive number of clinical trials targeting different antigens likely reflects the genetic heterogeneity of AML. The limited number of patients reported in multiple early clinical studies makes it difficult to draw conclusions about CAR safety, but it does suggest that the efficacy of this approach in AML lags behind the success observed in B cell malignancies. There is a clear need not only to improve CAR design but also to identify targets in AML that show limited expression in normal myeloid lineage cells.
Collapse
Affiliation(s)
- Swati Garg
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA 02215, USA; (W.N.); (J.D.G.); (M.S.)
- Department of Medicine, Harvard Medical School, Boston, MA 02115, USA
| | - Wei Ni
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA 02215, USA; (W.N.); (J.D.G.); (M.S.)
- Department of Medicine, Harvard Medical School, Boston, MA 02115, USA
| | - James D. Griffin
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA 02215, USA; (W.N.); (J.D.G.); (M.S.)
- Department of Medicine, Harvard Medical School, Boston, MA 02115, USA
| | - Martin Sattler
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA 02215, USA; (W.N.); (J.D.G.); (M.S.)
- Department of Medicine, Harvard Medical School, Boston, MA 02115, USA
| |
Collapse
|
48
|
Uscanga-Palomeque AC, Chávez-Escamilla AK, Alvizo-Báez CA, Saavedra-Alonso S, Terrazas-Armendáriz LD, Tamez-Guerra RS, Rodríguez-Padilla C, Alcocer-González JM. CAR-T Cell Therapy: From the Shop to Cancer Therapy. Int J Mol Sci 2023; 24:15688. [PMID: 37958672 PMCID: PMC10649325 DOI: 10.3390/ijms242115688] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2023] [Revised: 10/04/2023] [Accepted: 10/12/2023] [Indexed: 11/15/2023] Open
Abstract
Cancer is a worldwide health problem. Nevertheless, new technologies in the immunotherapy field have emerged. Chimeric antigen receptor (CAR) technology is a novel biological form to treat cancer; CAR-T cell genetic engineering has positively revolutionized cancer immunotherapy. In this paper, we review the latest developments in CAR-T in cancer treatment. We present the structure of the different generations and variants of CAR-T cells including TRUCK (T cells redirected for universal cytokine killing. We explain the approaches of the CAR-T cells manufactured ex vivo and in vivo. Moreover, we describe the limitations and areas of opportunity for this immunotherapy and the current challenges of treating hematological and solid cancer using CAR-T technology as well as its constraints and engineering approaches. We summarize other immune cells that have been using CAR technology, such as natural killer (NK), macrophages (M), and dendritic cells (DC). We conclude that CAR-T cells have the potential to treat not only cancer but other chronic diseases.
Collapse
Affiliation(s)
- Ashanti Concepción Uscanga-Palomeque
- Laboratorio de Inmunología y Virología, Facultad de Ciencias Biológicas, Universidad Autónoma de Nuevo León, San Nicolás de los Garza 66450, Nuevo León, Mexico; (A.K.C.-E.); (C.A.A.-B.); (S.S.-A.); (L.D.T.-A.); (R.S.T.-G.); (C.R.-P.)
| | | | | | | | | | | | | | - Juan Manuel Alcocer-González
- Laboratorio de Inmunología y Virología, Facultad de Ciencias Biológicas, Universidad Autónoma de Nuevo León, San Nicolás de los Garza 66450, Nuevo León, Mexico; (A.K.C.-E.); (C.A.A.-B.); (S.S.-A.); (L.D.T.-A.); (R.S.T.-G.); (C.R.-P.)
| |
Collapse
|
49
|
Lahimchi MR, Maroufi F, Maali A. Induced Pluripotent Stem Cell-Derived Chimeric Antigen Receptor T Cells: The Intersection of Stem Cells and Immunotherapy. Cell Reprogram 2023; 25:195-211. [PMID: 37782910 DOI: 10.1089/cell.2023.0041] [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] [Indexed: 10/04/2023] Open
Abstract
Chimeric antigen receptor (CAR) T cell therapy is a promising cell-based immunotherapy applicable to various cancers. High cost of production, immune rejection, heterogeneity of cell product, limited cell source, limited expandability, and relatively long production time have created the need to achieve a universal allogeneic CAR-T cell product for "off-the-shelf" application. Since the innovation of induced pluripotent stem cells (iPSCs) by Yamanaka et al., extensive efforts have been made to prepare an unlimited cell source for regenerative medicine, that is, immunotherapy. In the autologous grafting approach, iPSCs prepare the desired cell source for generating autologous CAR-T cells through more accessible and available sources. In addition, generating iPSC-derived CAR-T cells is a promising approach to achieving a suitable source for producing an allogeneic CAR-T cell product. In brief, the first step is reprogramming somatic cells (accessible from peripheral blood, skin, etc.) to iPSCs. In the next step, CAR expression and T cell lineage differentiation should be applied in different arrangements. In addition, in an allogeneic manner, human leukocyte antigen/T cell receptor (TCR) deficiency should be applied in iPSC colonies. The allogeneic iPSC-derived CAR-T cell experiments showed that simultaneous performance of HLA/TCR deficiency, CAR expression, and T cell lineage differentiation could bring the production to the highest efficacy in generating allogeneic iPSC-derived CAR-T cells.
Collapse
Affiliation(s)
| | - Faezeh Maroufi
- Department of Medical Laboratory Sciences, Faculty of Allied Medicine, Qazvin University of Medical Sciences, Qazvin, Iran
| | - Amirhosein Maali
- Department of Immunology, Pasteur Institute of Iran, Tehran, Iran
- Department of Medical Biotechnology, Faculty of Allied Medicine, Qazvin University of Medical Sciences, Qazvin, Iran
| |
Collapse
|
50
|
Alsajjan R, Mason WP. Bispecific T-Cell Engagers and Chimeric Antigen Receptor T-Cell Therapies in Glioblastoma: An Update. Curr Oncol 2023; 30:8501-8549. [PMID: 37754534 PMCID: PMC10529026 DOI: 10.3390/curroncol30090619] [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: 05/21/2023] [Revised: 09/04/2023] [Accepted: 09/08/2023] [Indexed: 09/28/2023] Open
Abstract
Glioblastoma is the most common malignant primary brain tumor in adults. The prognosis is extremely poor even with standard treatment of maximal safe resection, radiotherapy, and chemotherapy. Recurrence is inevitable within months, and treatment options are very limited. Chimeric antigen receptor T-cell therapy (CART) and bispecific T-cell engagers (TCEs) are two emerging immunotherapies that can redirect T-cells for tumor-specific killing and have shown remarkable success in hematological malignancies and been under extensive study for application in glioblastoma. While there have been multiple clinical trials showing preliminary evidence of safety and efficacy for CART, bispecific TCEs are still in the early stages of clinical testing, with preclinical studies showing very promising results. However, there are multiple shared challenges that need to be addressed in the future, including the route of delivery, antigen escape, the immunosuppressive tumor microenvironment, and toxicity resulting from the limited choice of tumor-specific antigens. Efforts are underway to optimize the design of both these treatments and find the ideal combination therapy to overcome these challenges. In this review, we describe the work that has been performed as well as novel approaches in glioblastoma and in other solid tumors that may be applicable in the future.
Collapse
Affiliation(s)
- Roa Alsajjan
- Division of Medical Oncology and Hematology, Princess Margaret Cancer Centre, University Health Network, University of Toronto, Toronto, ON M5G 2C1, Canada
- Division of Neurology, Department of Medicine, College of Medicine, King Saud University, Riyadh 11461, Saudi Arabia
| | - Warren P. Mason
- Division of Medical Oncology and Hematology, Princess Margaret Cancer Centre, University Health Network, University of Toronto, Toronto, ON M5G 2C1, Canada
| |
Collapse
|