1
|
Lim J, Oh D, Cheng M, Chintapula U, Liu S, Reynolds D, Zhang X, Zhou Y, Xu X, Ko J. Enhancing Chimeric Antigen Receptor T-Cell Generation via Microfluidic Mechanoporation and Lipid Nanoparticles. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2025; 21:e2410975. [PMID: 40103509 PMCID: PMC12036559 DOI: 10.1002/smll.202410975] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/17/2024] [Revised: 03/04/2025] [Indexed: 03/20/2025]
Abstract
Chimeric antigen receptor (CAR)-T cell therapy has revolutionized cancer treatment by engineering patients' T cells to specifically target cancer cells. Traditional CAR-T cell manufacturing methods use viral transduction to integrate CAR genes into T cells, but this can cause severe side effects and immune reactions and is costly. To overcome these challenges, non-viral methods, such as plasmid DNA (pDNA) transfection, are being explored. Here, a high-throughput intracellular delivery platform that integrates microfluidic mechanoporation with lipid nanoparticle (LNP)-based delivery, LNP + Squeeze, is introduced. This system enhances pDNA transfection efficiency in T cells while maintaining cell viability compared to other non-viral transfection methods like electroporation. This platform successfully engineers CAR-T cells using primary human T cells with a high transfection efficiency and demonstrates potent cytotoxicity against melanoma cells. This approach offers a promising, cost-effective, and scalable alternative to viral methods, potentially improving the accessibility and efficacy of CAR-T cell therapies.
Collapse
Affiliation(s)
- Jianhua Lim
- Department of BioengineeringUniversity of PennsylvaniaPhiladelphiaPA19104USA
| | - Daniel Oh
- Department of BioengineeringUniversity of PennsylvaniaPhiladelphiaPA19104USA
| | - Makayla Cheng
- Department of BioengineeringUniversity of PennsylvaniaPhiladelphiaPA19104USA
| | - Uday Chintapula
- Department of Pathology and Laboratory MedicinePerelman School of MedicineUniversity of PennsylvaniaPhiladelphiaPA19104USA
| | - Shujing Liu
- Department of Pathology and Laboratory MedicinePerelman School of MedicineUniversity of PennsylvaniaPhiladelphiaPA19104USA
| | - David Reynolds
- Department of BioengineeringUniversity of PennsylvaniaPhiladelphiaPA19104USA
| | - Xiaogang Zhang
- Department of Pathology and Laboratory MedicinePerelman School of MedicineUniversity of PennsylvaniaPhiladelphiaPA19104USA
| | - Yumeng Zhou
- Department of Pathology and Laboratory MedicinePerelman School of MedicineUniversity of PennsylvaniaPhiladelphiaPA19104USA
| | - Xiaowei Xu
- Department of Pathology and Laboratory MedicinePerelman School of MedicineUniversity of PennsylvaniaPhiladelphiaPA19104USA
| | - Jina Ko
- Department of BioengineeringUniversity of PennsylvaniaPhiladelphiaPA19104USA
- Department of Pathology and Laboratory MedicinePerelman School of MedicineUniversity of PennsylvaniaPhiladelphiaPA19104USA
| |
Collapse
|
2
|
Mosley SR, Chen A, Doell DNW, Choi S, Mowat C, Meier-Stephenson F, Meier-Stephenson V, Baker K. Cytosolic DNA composition is determined by genomic instability mechanism and regulates dendritic cell-mediated anti-tumor immunity. Cell Rep 2025; 44:115177. [PMID: 39864057 DOI: 10.1016/j.celrep.2024.115177] [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: 04/12/2024] [Revised: 11/10/2024] [Accepted: 12/18/2024] [Indexed: 01/28/2025] Open
Abstract
Patients with colorectal cancers (CRCs) that have microsatellite instability (MSI) (MSI CRCs) face a better prognosis than those with the more common chromosomal instability (CIN) subtype (CIN CRCs) due to improved T cell-mediated anti-tumor immune responses. Previous investigations identified the cytosolic DNA (cyDNA) sensor STING as necessary for chemokine-mediated T cell recruitment in MSI CRCs. Here, we find that cyDNA from MSI CRC cells is inherently more capable of inducing STING activation and improves cytotoxic T cell activation by dendritic cells (DCs). Sequencing indicates that MSI cyDNA is enriched in microsatellites, which, upon DC uptake, induce anti-tumor immunity in a manner consistent with clinical MSI CRCs. DNA-damaging therapies also modulate cyDNA stimulation capacity, with radiation inducing larger cyDNA sizes and increased mitochondrial DNA content. Identifying highly stimulatory endogenous cyDNAs such as those in MSI CRCs will allow for optimized development of DNA-based STING agonist therapies to improve the responses of CIN CRCs with CIN to immunotherapies.
Collapse
Affiliation(s)
- Shayla R Mosley
- Department of Oncology, Cross Cancer Institute, University of Alberta, Edmonton, AB T6G 1Z2, Canada.
| | - Angie Chen
- Department of Oncology, Cross Cancer Institute, University of Alberta, Edmonton, AB T6G 1Z2, Canada
| | - David N W Doell
- Department of Oncology, Cross Cancer Institute, University of Alberta, Edmonton, AB T6G 1Z2, Canada
| | - Siwon Choi
- Department of Oncology, Cross Cancer Institute, University of Alberta, Edmonton, AB T6G 1Z2, Canada
| | - Courtney Mowat
- Department of Oncology, Cross Cancer Institute, University of Alberta, Edmonton, AB T6G 1Z2, Canada
| | - Felix Meier-Stephenson
- Department of Medical Microbiology and Immunology, University of Alberta, Edmonton, AB T6G 2R3, Canada
| | - Vanessa Meier-Stephenson
- Department of Medical Microbiology and Immunology, University of Alberta, Edmonton, AB T6G 2R3, Canada
| | - Kristi Baker
- Department of Oncology, Cross Cancer Institute, University of Alberta, Edmonton, AB T6G 1Z2, Canada; Department of Medical Microbiology and Immunology, University of Alberta, Edmonton, AB T6G 2R3, Canada.
| |
Collapse
|
3
|
Chen Y, Chen X, Zhang Y, Wang M, Yang M, Wang R, Yan X, Shao S, Xin H, Hu Q, Wei W, Ping Y. Macrophage-specific in vivo RNA editing promotes phagocytosis and antitumor immunity in mice. Sci Transl Med 2025; 17:eadl5800. [PMID: 39813319 DOI: 10.1126/scitranslmed.adl5800] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2023] [Revised: 07/09/2024] [Accepted: 12/18/2024] [Indexed: 01/18/2025]
Abstract
Macrophages play a central role in antitumor immunity, making them an attractive target for gene therapy strategies. However, macrophages are difficult to transfect because of nucleic acid sensors that can trigger the degradation of foreign plasmid DNA. Here, we developed a macrophage-specific editing (MAGE) system by which compact plasmid DNA encoding a CasRx editor can be delivered to macrophages by a poly(β-amino ester) (PBAE) carrier to bypass the DNA sensor and enable RNA editing in vitro and in vivo. We identified a four-arm branched PBAE with 1-(2-aminoethyl)-4-methylpiperazine end-capping (PBAE29) that enables highly efficient macrophage transfection. PBAE29-mediated transfection of cultured macrophages stimulated less inflammatory cytokine production and inflammasome activation compared with traditional lipofectamine or electroporation-mediated plasmid delivery. Transfection efficiency was further improved by delivering CasRx by minicircle plasmid. The MAGE system incorporated a layer of carboxylated-mannan coating to target macrophage mannose receptors and a macrophage-specific promoter for enhanced selectivity. The delivery of CasRx with guide RNA targeting the transcripts for sialic acid-binding immunoglobulin similar to lectin 10 and signal regulatory protein alpha expression resulted in effective protein knockdown, improving macrophage phagocytosis. The MAGE system also showed efficacy in targeting macrophages in vivo, stimulating antitumor immune responses and reducing tumor volume in murine tumor models, including patient-derived pancreatic adenocarcinoma xenografts in humanized mice. In sum, the MAGE system presents a promising platform for in vivo macrophage-specific delivery of RNA editing tools that can be applied as a cancer therapy.
Collapse
Affiliation(s)
- Yuxuan Chen
- College of Pharmaceutical Sciences, State Key Laboratory of Advanced Drug Delivery and Release Systems, Zhejiang University, Hangzhou 310058, China
- Liangzhu Laboratory, Zhejiang University, Hangzhou 311121, China
| | - Xiaohong Chen
- College of Pharmaceutical Sciences, State Key Laboratory of Advanced Drug Delivery and Release Systems, Zhejiang University, Hangzhou 310058, China
- Liangzhu Laboratory, Zhejiang University, Hangzhou 311121, China
- Zhejiang Provincial Key Laboratory of Pancreatic Disease, MOE Joint International Research Laboratory of Pancreatic Diseases, First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310003, China
| | - Yao Zhang
- College of Pharmaceutical Sciences, State Key Laboratory of Advanced Drug Delivery and Release Systems, Zhejiang University, Hangzhou 310058, China
| | - Meng Wang
- Zhejiang Provincial Key Laboratory of Pancreatic Disease, MOE Joint International Research Laboratory of Pancreatic Diseases, First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310003, China
| | - Minqi Yang
- Zhejiang Provincial Key Laboratory of Pancreatic Disease, MOE Joint International Research Laboratory of Pancreatic Diseases, First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310003, China
| | - Ruiji Wang
- College of Pharmaceutical Sciences, State Key Laboratory of Advanced Drug Delivery and Release Systems, Zhejiang University, Hangzhou 310058, China
| | - Xiaojie Yan
- College of Pharmaceutical Sciences, State Key Laboratory of Advanced Drug Delivery and Release Systems, Zhejiang University, Hangzhou 310058, China
- Liangzhu Laboratory, Zhejiang University, Hangzhou 311121, China
| | - Shiyi Shao
- Zhejiang Provincial Key Laboratory of Pancreatic Disease, MOE Joint International Research Laboratory of Pancreatic Diseases, First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310003, China
| | - Huhu Xin
- College of Pharmaceutical Sciences, State Key Laboratory of Advanced Drug Delivery and Release Systems, Zhejiang University, Hangzhou 310058, China
| | - Qida Hu
- Zhejiang Provincial Key Laboratory of Pancreatic Disease, MOE Joint International Research Laboratory of Pancreatic Diseases, First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310003, China
| | - Wei Wei
- State Key Laboratory of Biochemical Engineering, Institute of Process Engineering, Chinese Academy of Sciences, Beijing 100190, China
- School of Chemical Engineering, University of Chinese Academy of Sciences, Beijing 100049, China
| | - Yuan Ping
- College of Pharmaceutical Sciences, State Key Laboratory of Advanced Drug Delivery and Release Systems, Zhejiang University, Hangzhou 310058, China
- Liangzhu Laboratory, Zhejiang University, Hangzhou 311121, China
| |
Collapse
|
4
|
Park SJ, Kweon S, Moyo MK, Kim HR, Choi JU, Lee NK, Maharjan R, Cho YS, Park JW, Byun Y. Immune modulation of the liver metastatic colorectal cancer microenvironment via the oral CAPOX-mediated cGAS-STING pathway. Biomaterials 2024; 310:122625. [PMID: 38820768 DOI: 10.1016/j.biomaterials.2024.122625] [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: 01/30/2024] [Revised: 05/05/2024] [Accepted: 05/19/2024] [Indexed: 06/02/2024]
Abstract
We evaluated modulation of the immunosuppressive tumor microenvironment in both local and liver metastatic colorectal cancer (LMCC), focusing on tumor-associated macrophages, which are the predominant immunosuppressive cells in LMCC. We developed an orally administered metronomic chemotherapy regimen, oral CAPOX. This regimen combines capecitabine and a nano-micelle encapsulated, lysine-linked deoxycholate and oxaliplatin complex (OPt/LDC-NM). The treatment effectively modulated immune cells within the tumor microenvironment by activating the cGAS-STING pathway and inducing immunogenic cell death. This therapy modulated immune cells more effectively than did capecitabine monotherapy, the current standard maintenance chemotherapy for colorectal cancer. The macrophage-modifying effect of oral CAPOX was mediated via the cGAS-STING pathway. This is a newly identified mode of immune cell activation induced by metronomic chemotherapy. Moreover, oral CAPOX synergized with anti-PD-1 antibody (αPD-1) to enhance the T-cell-mediated antitumor immune response. In the CT26. CL25 subcutaneous model, combination therapy achieved a 91 % complete response rate with a confirmed memory effect against the tumor. This combination also altered the immunosuppressive tumor microenvironment in LMCC, which αPD-1 monotherapy could not achieve. Oral CAPOX and αPD-1 combination therapy outperformed the maximum tolerated dose for treating LMCC, suggesting metronomic therapy as a promising strategy.
Collapse
Affiliation(s)
- Seong Jin Park
- Research Institute of Pharmaceutical Sciences, College of Pharmacy, Seoul National University, Seoul 08826, Republic of Korea
| | - Seho Kweon
- College of Pharmacy, Chonnam National University, Gwangju 61186, Republic of Korea
| | | | - Ha Rin Kim
- Research Institute of Pharmaceutical Sciences, College of Pharmacy, Seoul National University, Seoul 08826, Republic of Korea; School of Medicine, Oncology, Stanford University, CA, 94305, United States
| | - Jeong Uk Choi
- College of Pharmacy, Kyung Hee University, Dongdaemun-gu, Seoul, Republic of Korea
| | - Na Kyeong Lee
- College of Pharmacy and Research Institute for Drug Development, Pusan National University, Busan, 46241, Republic of Korea
| | - Ruby Maharjan
- Massachusetts General Hospital Cancer Center, Department of Medicine, Harvard Medical School, Boston, MA 02114, United States
| | - Young Seok Cho
- College of Pharmacy, University of Michigan, Ann Arbor, MI, United States
| | - Jin Woo Park
- College of Pharmacy and Natural Medicine Research Institute, Mokpo National University, Jeonnam 58554, Republic of Korea; Department of Biomedicine, Health & Life Convergence Sciences, BK21 Four, Biomedical and Healthcare Research Institute, Mokpo National University, Jeonnam 58554, Republic of Korea.
| | - Youngro Byun
- Research Institute of Pharmaceutical Sciences, College of Pharmacy, Seoul National University, Seoul 08826, Republic of Korea.
| |
Collapse
|
5
|
Hodgins JJ, Abou-Hamad J, O’Dwyer CE, Hagerman A, Yakubovich E, Tanese de Souza C, Marotel M, Buchler A, Fadel S, Park MM, Fong-McMaster C, Crupi MF, Makinson OJ, Kurdieh R, Rezaei R, Dhillon HS, Ilkow CS, Bell JC, Harper ME, Rotstein BH, Auer RC, Vanderhyden BC, Sabourin LA, Bourgeois-Daigneault MC, Cook DP, Ardolino M. PD-L1 promotes oncolytic virus infection via a metabolic shift that inhibits the type I IFN pathway. J Exp Med 2024; 221:e20221721. [PMID: 38869480 PMCID: PMC11176258 DOI: 10.1084/jem.20221721] [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: 10/11/2022] [Revised: 02/04/2024] [Accepted: 03/14/2024] [Indexed: 06/14/2024] Open
Abstract
While conventional wisdom initially postulated that PD-L1 serves as the inert ligand for PD-1, an emerging body of literature suggests that PD-L1 has cell-intrinsic functions in immune and cancer cells. In line with these studies, here we show that engagement of PD-L1 via cellular ligands or agonistic antibodies, including those used in the clinic, potently inhibits the type I interferon pathway in cancer cells. Hampered type I interferon responses in PD-L1-expressing cancer cells resulted in enhanced efficacy of oncolytic viruses in vitro and in vivo. Consistently, PD-L1 expression marked tumor explants from cancer patients that were best infected by oncolytic viruses. Mechanistically, PD-L1 promoted a metabolic shift characterized by enhanced glycolysis rate that resulted in increased lactate production. In turn, lactate inhibited type I IFN responses. In addition to adding mechanistic insight into PD-L1 intrinsic function, our results will also help guide the numerous ongoing efforts to combine PD-L1 antibodies with oncolytic virotherapy in clinical trials.
Collapse
Affiliation(s)
- Jonathan J. Hodgins
- Cancer Therapeutics Program, Ottawa Hospital Research Institute, Ottawa, Canada
- Department of Biochemistry, Microbiology, and Immunology, University of Ottawa, Ottawa, Canada
- Center for Infection, Immunity, and Inflammation, University of Ottawa, Ottawa, Canada
| | - John Abou-Hamad
- Cancer Therapeutics Program, Ottawa Hospital Research Institute, Ottawa, Canada
- Department of Cellular and Molecular Medicine, University of Ottawa, Ottawa, Canada
| | - Colin Edward O’Dwyer
- Cancer Therapeutics Program, Ottawa Hospital Research Institute, Ottawa, Canada
- Department of Biochemistry, Microbiology, and Immunology, University of Ottawa, Ottawa, Canada
- Center for Infection, Immunity, and Inflammation, University of Ottawa, Ottawa, Canada
| | - Ash Hagerman
- Cancer Therapeutics Program, Ottawa Hospital Research Institute, Ottawa, Canada
- Center for Infection, Immunity, and Inflammation, University of Ottawa, Ottawa, Canada
| | - Edward Yakubovich
- Cancer Therapeutics Program, Ottawa Hospital Research Institute, Ottawa, Canada
- Department of Cellular and Molecular Medicine, University of Ottawa, Ottawa, Canada
| | | | - Marie Marotel
- Cancer Therapeutics Program, Ottawa Hospital Research Institute, Ottawa, Canada
- Center for Infection, Immunity, and Inflammation, University of Ottawa, Ottawa, Canada
| | - Ariel Buchler
- Department of Chemistry and Biomolecular Sciences, University of Ottawa, Ottawa, Canada
- University of Ottawa Heart Institute, Ottawa, Canada
| | - Saleh Fadel
- The Ottawa Hospital, Ottawa, Canada
- Department of Pathology and Laboratory Medicine, The Ottawa Hospital, Ottawa, Canada
| | - Maria M. Park
- Cancer Therapeutics Program, Ottawa Hospital Research Institute, Ottawa, Canada
- Center for Infection, Immunity, and Inflammation, University of Ottawa, Ottawa, Canada
| | - Claire Fong-McMaster
- Department of Biochemistry, Microbiology, and Immunology, University of Ottawa, Ottawa, Canada
- Ottawa Institute for Systems Biology, Ottawa, Canada
| | - Mathieu F. Crupi
- Cancer Therapeutics Program, Ottawa Hospital Research Institute, Ottawa, Canada
| | - Olivia Joan Makinson
- Cancer Therapeutics Program, Ottawa Hospital Research Institute, Ottawa, Canada
- Department of Biochemistry, Microbiology, and Immunology, University of Ottawa, Ottawa, Canada
- Center for Infection, Immunity, and Inflammation, University of Ottawa, Ottawa, Canada
| | - Reem Kurdieh
- Cancer Therapeutics Program, Ottawa Hospital Research Institute, Ottawa, Canada
| | - Reza Rezaei
- Cancer Therapeutics Program, Ottawa Hospital Research Institute, Ottawa, Canada
- Department of Biochemistry, Microbiology, and Immunology, University of Ottawa, Ottawa, Canada
- Center for Infection, Immunity, and Inflammation, University of Ottawa, Ottawa, Canada
| | - Harkirat Singh Dhillon
- Cancer Therapeutics Program, Ottawa Hospital Research Institute, Ottawa, Canada
- Department of Biochemistry, Microbiology, and Immunology, University of Ottawa, Ottawa, Canada
- Center for Infection, Immunity, and Inflammation, University of Ottawa, Ottawa, Canada
| | - Carolina S. Ilkow
- Cancer Therapeutics Program, Ottawa Hospital Research Institute, Ottawa, Canada
- Department of Biochemistry, Microbiology, and Immunology, University of Ottawa, Ottawa, Canada
- Center for Infection, Immunity, and Inflammation, University of Ottawa, Ottawa, Canada
| | - John C. Bell
- Cancer Therapeutics Program, Ottawa Hospital Research Institute, Ottawa, Canada
- Department of Biochemistry, Microbiology, and Immunology, University of Ottawa, Ottawa, Canada
| | - Mary-Ellen Harper
- Department of Biochemistry, Microbiology, and Immunology, University of Ottawa, Ottawa, Canada
- Center for Infection, Immunity, and Inflammation, University of Ottawa, Ottawa, Canada
- Ottawa Institute for Systems Biology, Ottawa, Canada
| | - Benjamin H. Rotstein
- Department of Biochemistry, Microbiology, and Immunology, University of Ottawa, Ottawa, Canada
- Department of Chemistry and Biomolecular Sciences, University of Ottawa, Ottawa, Canada
- University of Ottawa Heart Institute, Ottawa, Canada
| | - Rebecca C. Auer
- Cancer Therapeutics Program, Ottawa Hospital Research Institute, Ottawa, Canada
- Department of Biochemistry, Microbiology, and Immunology, University of Ottawa, Ottawa, Canada
| | - Barbara C. Vanderhyden
- Cancer Therapeutics Program, Ottawa Hospital Research Institute, Ottawa, Canada
- Center for Infection, Immunity, and Inflammation, University of Ottawa, Ottawa, Canada
- Department of Cellular and Molecular Medicine, University of Ottawa, Ottawa, Canada
| | - Luc A. Sabourin
- Cancer Therapeutics Program, Ottawa Hospital Research Institute, Ottawa, Canada
- Department of Cellular and Molecular Medicine, University of Ottawa, Ottawa, Canada
| | - Marie-Claude Bourgeois-Daigneault
- Department of Microbiology, Infectious Diseases, and Immunology, University of Montreal, Montreal, Canada
- Centre Hospitalier de l’Université de Montréal Research Centre, Cancer and Immunopathology axes, Montreal, Canada
| | - David P. Cook
- Cancer Therapeutics Program, Ottawa Hospital Research Institute, Ottawa, Canada
- Department of Cellular and Molecular Medicine, University of Ottawa, Ottawa, Canada
| | - Michele Ardolino
- Cancer Therapeutics Program, Ottawa Hospital Research Institute, Ottawa, Canada
- Department of Biochemistry, Microbiology, and Immunology, University of Ottawa, Ottawa, Canada
- Center for Infection, Immunity, and Inflammation, University of Ottawa, Ottawa, Canada
| |
Collapse
|
6
|
Cornista AM, Giolito MV, Baker K, Hazime H, Dufait I, Datta J, Khumukcham SS, De Ridder M, Roper J, Abreu MT, Breckpot K, Van der Jeught K. Colorectal Cancer Immunotherapy: State of the Art and Future Directions. GASTRO HEP ADVANCES 2023; 2:1103-1119. [PMID: 38098742 PMCID: PMC10721132 DOI: 10.1016/j.gastha.2023.09.007] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/17/2023]
Abstract
Cancer immunotherapy has become an indispensable mode of treatment for a multitude of solid tumor cancers. Colorectal cancer (CRC) has been one of the many cancer types to benefit from immunotherapy, especially in advanced disease where standard treatment fails to prevent recurrence or results in poor survival. The efficacy of immunotherapy in CRC has not been without challenge, as early clinical trials observed dismal responses in unselected CRC patients treated with checkpoint inhibitors. Many studies and clinical trials have since refined immunotherapies available for CRC, solidifying immunotherapy as a powerful asset for CRC treatment. This review article examines CRC immunotherapies, from their foundation, through emerging avenues for improvement, to future directions.
Collapse
Affiliation(s)
- Alyssa Mauri Cornista
- Department of Microbiology and Immunology, University of Miami Miller School of Medicine, Miami, Florida
| | - Maria Virginia Giolito
- Department of Biomedical Sciences, Vrije Universiteit Brussel, Laboratory for Molecular and Cellular Therapy, Brussels, Belgium
| | - Kristi Baker
- Department of Oncology, University of Alberta, Edmonton, Canada
- Department of Medical Microbiology and Immunology, University of Alberta, Edmonton, Canada
| | - Hajar Hazime
- Division of Gastroenterology, University of Miami Miller School of Medicine, Miami, Florida
| | - Inès Dufait
- Department of Radiotherapy, Universitair Ziekenhuis Brussel, Vrije Universiteit Brussel, Brussels, Belgium
| | - Jashodeep Datta
- Sylvester Comprehensive Cancer Center, University of Miami, Miami, Florida
- Division of Surgical Oncology, Dewitt Daughtry Department of Surgery, University of Miami Miller School of Medicine, Miami, Florida
| | - Saratchandra Singh Khumukcham
- Division of Gastroenterology, Department of Medicine, Duke University, Durham, North Carolina
- Department of Pharmacology and Cancer Biology, Duke University, Durham, North Carolina
| | - Mark De Ridder
- Department of Radiotherapy, Universitair Ziekenhuis Brussel, Vrije Universiteit Brussel, Brussels, Belgium
| | - Jatin Roper
- Division of Gastroenterology, Department of Medicine, Duke University, Durham, North Carolina
- Department of Pharmacology and Cancer Biology, Duke University, Durham, North Carolina
| | - Maria T. Abreu
- Division of Gastroenterology, University of Miami Miller School of Medicine, Miami, Florida
- Sylvester Comprehensive Cancer Center, University of Miami, Miami, Florida
| | - Karine Breckpot
- Department of Biomedical Sciences, Vrije Universiteit Brussel, Laboratory for Molecular and Cellular Therapy, Brussels, Belgium
| | - Kevin Van der Jeught
- Department of Microbiology and Immunology, University of Miami Miller School of Medicine, Miami, Florida
- Sylvester Comprehensive Cancer Center, University of Miami, Miami, Florida
| |
Collapse
|
7
|
Wahl D, Smith ME, McEntee CM, Cavalier AN, Osburn SC, Burke SD, Grant RA, Nerguizian D, Lark DS, Link CD, LaRocca TJ. The reverse transcriptase inhibitor 3TC protects against age-related cognitive dysfunction. Aging Cell 2023; 22:e13798. [PMID: 36949552 PMCID: PMC10186603 DOI: 10.1111/acel.13798] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2022] [Revised: 01/30/2023] [Accepted: 02/01/2023] [Indexed: 03/24/2023] Open
Abstract
Aging is the primary risk factor for most neurodegenerative diseases, including Alzheimer's disease. Major hallmarks of brain aging include neuroinflammation/immune activation and reduced neuronal health/function. These processes contribute to cognitive dysfunction (a key risk factor for Alzheimer's disease), but their upstream causes are incompletely understood. Age-related increases in transposable element (TE) transcripts might contribute to reduced cognitive function with brain aging, as the reverse transcriptase inhibitor 3TC reduces inflammation in peripheral tissues and TE transcripts have been linked with tau pathology in Alzheimer's disease. However, the effects of 3TC on cognitive function with aging have not been investigated. Here, in support of a role for TE transcripts in brain aging/cognitive decline, we show that 3TC: (a) improves cognitive function and reduces neuroinflammation in old wild-type mice; (b) preserves neuronal health with aging in mice and Caenorhabditis elegans; and (c) enhances cognitive function in a mouse model of tauopathy. We also provide insight on potential underlying mechanisms, as well as evidence of translational relevance for these observations by showing that TE transcripts accumulate with brain aging in humans, and that these age-related increases intersect with those observed in Alzheimer's disease. Collectively, our results suggest that TE transcript accumulation during aging may contribute to cognitive decline and neurodegeneration, and that targeting these events with reverse transcriptase inhibitors like 3TC could be a viable therapeutic strategy.
Collapse
Affiliation(s)
- Devin Wahl
- Department of Health and Exercise ScienceColorado State UniversityFort CollinsColoradoUSA
- Center for Healthy AgingColorado State UniversityFort CollinsColoradoUSA
| | - Meghan E. Smith
- Department of Health and Exercise ScienceColorado State UniversityFort CollinsColoradoUSA
- Center for Healthy AgingColorado State UniversityFort CollinsColoradoUSA
| | - Cali M. McEntee
- Department of Health and Exercise ScienceColorado State UniversityFort CollinsColoradoUSA
- Center for Healthy AgingColorado State UniversityFort CollinsColoradoUSA
| | - Alyssa N. Cavalier
- Department of Health and Exercise ScienceColorado State UniversityFort CollinsColoradoUSA
- Center for Healthy AgingColorado State UniversityFort CollinsColoradoUSA
| | - Shelby C. Osburn
- Department of Health and Exercise ScienceColorado State UniversityFort CollinsColoradoUSA
- Center for Healthy AgingColorado State UniversityFort CollinsColoradoUSA
| | - Samuel D. Burke
- Department of Health and Exercise ScienceColorado State UniversityFort CollinsColoradoUSA
- Center for Healthy AgingColorado State UniversityFort CollinsColoradoUSA
| | - Randy A. Grant
- Department of Health and Exercise ScienceColorado State UniversityFort CollinsColoradoUSA
- Center for Healthy AgingColorado State UniversityFort CollinsColoradoUSA
| | - David Nerguizian
- Department of Biochemistry and Molecular GeneticsUniversity of Colorado School of MedicineAuroraColoradoUSA
| | - Daniel S. Lark
- Department of Health and Exercise ScienceColorado State UniversityFort CollinsColoradoUSA
| | - Christopher D. Link
- Department of Integrative PhysiologyUniversity of Colorado BoulderBoulderColoradoUSA
| | - Thomas J. LaRocca
- Department of Health and Exercise ScienceColorado State UniversityFort CollinsColoradoUSA
- Center for Healthy AgingColorado State UniversityFort CollinsColoradoUSA
| |
Collapse
|