1
|
Smith KWY, Fung SL, Wu HF, Chiesa I, Vozzi G, De Maria C, Gottardi R. Developing an in vitro osteochondral micro-physiological system for modeling cartilage-bone crosstalk in arthritis. Front Immunol 2025; 16:1495613. [PMID: 40491903 PMCID: PMC12146386 DOI: 10.3389/fimmu.2025.1495613] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2024] [Accepted: 04/10/2025] [Indexed: 06/11/2025] Open
Abstract
Introduction Arthritis, a disease affecting over 50 million adults in the United States, encompasses many different conditions involving joints and surrounding tissues. Disease development, progression, and subsequent treatment is dependent on many different factors, including the relationship between adjacent tissues and the immunological signals involved. A major contributor to disease regulation is the crosstalk between the cartilage and the bone in joints, as well as their reaction to immune factors such as cytokine signaling and macrophage mediation. Studying cartilage-bone crosstalk in arthritis development can be difficult, as controlling immunological factors in vivo is challenging, but in vitro models often lack multi-tissue relevancy. Methods To fix this, we developed an in vitro micro-physiological system using a biphasic bioreactor that supports modeling of multiple tissues. We generated cartilage and vascularized-bone analogs and combined them in the bioreactor to allow diffusion and signaling between them. Using this system, we directly induced inflammation in the cartilage region and studied how crosstalk between the two adjacent tissues contributed to disease progression. Results We showed that conditioned media from pro-inflammatory macrophages generated a different inflammatory profile than a simple inflammatory cytokine cocktail. We also showed that the vascularized-bone region became inflamed in response to the cartilage inflammation, verifying crosstalk in the system and successfully modeling the relationship between cartilage and bone in an arthritic environment. Discussion This model can be used to further probe the crosstalk between bone and cartilage in arthritis, allowing researchers to tease out the effect of specific inflammatory agents or therapeutics in vitro.
Collapse
Affiliation(s)
- Kyra W. Y. Smith
- Department of Bioengineering, School of Engineering and Applied Science, University of Pennsylvania, Philadelphia, PA, United States
- Pulmonary and Sleep Medicine, Children’s Hospital of Philadelphia, Philadelphia, PA, United States
| | - Stephanie L. Fung
- Pulmonary and Sleep Medicine, Children’s Hospital of Philadelphia, Philadelphia, PA, United States
| | - Hsin-Fang Wu
- Department of Bioengineering, School of Engineering and Applied Science, University of Pennsylvania, Philadelphia, PA, United States
- Pulmonary and Sleep Medicine, Children’s Hospital of Philadelphia, Philadelphia, PA, United States
| | - Irene Chiesa
- Pulmonary and Sleep Medicine, Children’s Hospital of Philadelphia, Philadelphia, PA, United States
- Department of Information Engineering and Research Center E. Piaggio, University of Pisa, Pisa, Italy
| | - Giovanni Vozzi
- Department of Information Engineering and Research Center E. Piaggio, University of Pisa, Pisa, Italy
| | - Carmelo De Maria
- Department of Information Engineering and Research Center E. Piaggio, University of Pisa, Pisa, Italy
| | - Riccardo Gottardi
- Department of Bioengineering, School of Engineering and Applied Science, University of Pennsylvania, Philadelphia, PA, United States
- Pulmonary and Sleep Medicine, Children’s Hospital of Philadelphia, Philadelphia, PA, United States
- Division of Otolaryngology, Department of Surgery, Children's Hospital of Philadelphia, Philadelphia, PA, United States
- Department of Otorhinolaryngology, Head and Neck Surgery, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, United States
- Department of Orthopaedic Surgery, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA, United States
- Ri.MED Foundation, Palermo, PA, Italy
| |
Collapse
|
2
|
Bougnères P, Le Stunff C. Revisiting the Pathogenesis of X-Linked Adrenoleukodystrophy. Genes (Basel) 2025; 16:590. [PMID: 40428412 PMCID: PMC12111468 DOI: 10.3390/genes16050590] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2025] [Revised: 05/11/2025] [Accepted: 05/15/2025] [Indexed: 05/29/2025] Open
Abstract
BACKGROUND X-ALD is a white matter (WM) disease caused by mutations in the ABCD1 gene encoding the transporter of very-long-chain fatty acids (VLCFAs) into peroxisomes. Strikingly, the same ABCD1 mutation causes either devastating brain inflammatory demyelination during childhood or, more often, progressive spinal cord axonopathy starting in middle-aged adults. The accumulation of undegraded VLCFA in glial cell membranes and myelin has long been thought to be the central mechanism of X-ALD. METHODS This review discusses studies in mouse and drosophila models that have modified our views of X-ALD pathogenesis. RESULTS In the Abcd1 knockout (KO) mouse that mimics the spinal cord disease, the late manifestations of axonopathy are rapidly reversed by ABCD1 gene transfer into spinal cord oligodendrocytes (OLs). In a peroxin-5 KO mouse model, the selective impairment of peroxisomal biogenesis in OLs achieves an almost perfect phenocopy of cerebral ALD. A drosophila knockout model revealed that VLCFA accumulation in glial myelinating cells causes the production of a toxic lipid able to poison axons and activate inflammatory cells. Other mouse models showed the critical role of OLs in providing energy substrates to axons. In addition, studies on microglial changing substates have improved our understanding of neuroinflammation. CONCLUSIONS Animal models supporting a primary role of OLs and axonal pathology and a secondary role of microglia allow us to revisit of X-ALD mechanisms. Beyond ABCD1 mutations, pathogenesis depends on unidentified contributors, such as genetic background, cell-specific epigenomics, potential environmental triggers, and stochasticity of crosstalk between multiple cell types among billions of glial cells and neurons.
Collapse
Affiliation(s)
- Pierre Bougnères
- MIRCen Institute, Commissariat à l’Energie Atomique, Laboratoire des Maladies Neurodégénératives, 92260 Fontenay-aux-Roses, France
- NEURATRIS, 92260 Fontenay-aux-Roses, France
- Therapy Design Consulting, 94300 Vincennes, France
| | - Catherine Le Stunff
- MIRCen Institute, Commissariat à l’Energie Atomique, Laboratoire des Maladies Neurodégénératives, 92260 Fontenay-aux-Roses, France
- NEURATRIS, 92260 Fontenay-aux-Roses, France
- UMR1195 Inserm, University Paris Saclay, 94270 Le Kremlin-Bicêtre, France
| |
Collapse
|
3
|
Yan X, Arcoverde Cerveira R, Ols S, Lenart K, Hellgren F, Miranda M, Engstrand O, Reinhardt A, Eriksson B, Loré K. Biochemical and hematological reference intervals in rhesus and cynomolgus macaques and implications for vaccine and drug development. Lab Anim (NY) 2025:10.1038/s41684-025-01547-y. [PMID: 40379874 DOI: 10.1038/s41684-025-01547-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2024] [Accepted: 03/26/2025] [Indexed: 05/19/2025]
Abstract
Nonhuman primates have a key role in the evaluation of novel therapeutics including vaccine and drug development. Monitoring biochemical and hematological parameters of macaques is critical to understand toxicity and safety, but general reference intervals following standardized guidelines remain to be determined. Here we compiled multiple internal datasets to define normal ranges of classical biochemical and hematological parameters in Indian and Chinese rhesus macaques as well as cynomolgus macaques. Furthermore, the combination of hematological data with phenotypic information of cells obtained by flow cytometry enabled analyses of specific immune cell subsets. We found that vaccination generally induced transient changes at 24 h in cell frequencies accompanied by fluctuation in selected liver enzymes and metabolites. However, most parameters remained within our identified reference intervals. These deviations did not lead to noticeable side effects. Fluctuation in selected biochemical and hematological parameters was accompanied with differentiation of CD14+CD16+ intermediate monocytes and upregulation of genes associated with interleukin-1 signaling. By contrast, two animals with noticeable side effects showed sustained deviations. This study provides insights into baseline and vaccine-induced biochemical and hematological profiles of healthy macaques, facilitating the interpretation of toxicity and safety assessments in preclinical trials of novel therapies.
Collapse
Affiliation(s)
- Xianglei Yan
- Division of Immunology and Respiratory Medicine, Department of Medicine Solna, Karolinska Institutet and Karolinska University Hospital, Stockholm, Sweden
- Center of Molecular Medicine, Stockholm, Sweden
| | - Rodrigo Arcoverde Cerveira
- Division of Immunology and Respiratory Medicine, Department of Medicine Solna, Karolinska Institutet and Karolinska University Hospital, Stockholm, Sweden
- Center of Molecular Medicine, Stockholm, Sweden
| | - Sebastian Ols
- Division of Immunology and Respiratory Medicine, Department of Medicine Solna, Karolinska Institutet and Karolinska University Hospital, Stockholm, Sweden
- Center of Molecular Medicine, Stockholm, Sweden
- Institute for Protein Design, University of Washington, Seattle, WA, USA
| | - Klara Lenart
- Division of Immunology and Respiratory Medicine, Department of Medicine Solna, Karolinska Institutet and Karolinska University Hospital, Stockholm, Sweden
- Center of Molecular Medicine, Stockholm, Sweden
| | - Fredrika Hellgren
- Division of Immunology and Respiratory Medicine, Department of Medicine Solna, Karolinska Institutet and Karolinska University Hospital, Stockholm, Sweden
- Center of Molecular Medicine, Stockholm, Sweden
| | - Marcos Miranda
- Division of Immunology and Respiratory Medicine, Department of Medicine Solna, Karolinska Institutet and Karolinska University Hospital, Stockholm, Sweden
- Center of Molecular Medicine, Stockholm, Sweden
| | - Olivia Engstrand
- Division of Immunology and Respiratory Medicine, Department of Medicine Solna, Karolinska Institutet and Karolinska University Hospital, Stockholm, Sweden
- Center of Molecular Medicine, Stockholm, Sweden
| | - Annika Reinhardt
- Division of Immunology and Respiratory Medicine, Department of Medicine Solna, Karolinska Institutet and Karolinska University Hospital, Stockholm, Sweden
- Center of Molecular Medicine, Stockholm, Sweden
| | - Bengt Eriksson
- Astrid Fagraeus Laboratory, Comparative Medicine, Karolinska Institutet, Stockholm, Sweden
| | - Karin Loré
- Division of Immunology and Respiratory Medicine, Department of Medicine Solna, Karolinska Institutet and Karolinska University Hospital, Stockholm, Sweden.
- Center of Molecular Medicine, Stockholm, Sweden.
| |
Collapse
|
4
|
de Moura IA, Silva AJD, de Macêdo LS, de Melo KMTB, Leal LRS, Espinoza BCF, Invenção MDCV, de Pinho SS, de Freitas AC. Advances in the Functionalization of Vaccine Delivery Systems: Innovative Strategies and Translational Perspectives. Pharmaceutics 2025; 17:640. [PMID: 40430931 PMCID: PMC12115142 DOI: 10.3390/pharmaceutics17050640] [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: 03/31/2025] [Revised: 04/28/2025] [Accepted: 05/06/2025] [Indexed: 05/29/2025] Open
Abstract
The development of effective vaccines requires a rational design that considers the interaction between antigens, their vectors, and the immune system in addition to the activation of pathways that induce a safe and specific immune response. The efficacy of a vaccine formulation depends on the nature of the antigen, the protection offered by the delivery system, the ability to potentiate the immune response, and the precise release of the immunogen. Carrier systems such as lipid nanoparticles, polymers, exosomes, and microorganisms can be functionalized by chemical, physical, or biological methods to generate selective and improved biodistribution profiles. These methods enhance interaction with target cells, thereby improving immunological efficacy. The conjugation of specific ligands or the modification of parameters such as shape, charge, and size of vectors can enhance the specificity, stability, and efficiency of antigen transport to cellular compartments, thereby facilitating a robust immune response. This study examines modifications in vaccine delivery systems, focusing on biomolecules and physicochemical changes that enhance antigen presentation. Additionally, we examine innovative methods, including microneedles, electroporation, and needle-free systems that show potential for enhancing the immune response.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | - Antonio Carlos de Freitas
- Laboratory of Molecular Studies and Experimental Therapy—LEMTE, Department of Genetics, Federal University of Pernambuco, Avenida da Engenharia S/N, Recife 50740-600, Pernambuco, Brazil; (I.A.d.M.); (A.J.D.S.); (L.S.d.M.); (K.M.T.B.d.M.); (L.R.S.L.); (B.C.F.E.); (M.d.C.V.I.); (S.S.d.P.)
| |
Collapse
|
5
|
Moawad F, Pouliot R, Brambilla D. Dissolving microneedles in transdermal drug delivery: A critical analysis of limitations and translation challenges. J Control Release 2025:113794. [PMID: 40319916 DOI: 10.1016/j.jconrel.2025.113794] [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: 02/19/2025] [Revised: 04/25/2025] [Accepted: 04/28/2025] [Indexed: 05/07/2025]
Abstract
Microneedles (MNs) have emerged as an innovative approach for transdermal drug delivery, offering an efficient and minimally invasive alternative to conventional injections and oral delivery systems. While their potential has been widely recognized and extensively studied, the translation of MN technology into clinical practice remains limited. Despite the vast amount of published research, much of it involves over-complexification without addressing the core barriers to practical application. For example, dissolving/degradable MNs face key limitations such as poor drug loading capacity, low dosing consistency, and challenges in delivering effective therapeutic concentrations. These constraints restrict their utility to niche applications, such as vaccination or delivering potent drugs that require minimal doses. Additionally, the lack of standardized quality control measures, the complex manufacturing processes, and the high costs associated specifically with sterile/aseptic production further impede clinical translation. Regulatory frameworks for MNs remain vague, slowing the development of products that meet approval standards. This review critically examines the fundamental barriers to dissolving/degradable MN commercialization, as the most studied type of MN, while exploring promising strategies to overcome them. Advances in formulation science, fabrication techniques, and material engineering have demonstrated potential in enhancing drug loading efficiency and delivery consistency. Moreover, the establishment of clearer regulatory guidelines and scalable production strategies could significantly accelerate the commercialization of MN technology. By shifting focus toward pragmatic and clinically relevant solutions, this review aims to bridge the gap between research innovations and real-world applications, paving the way for broader implementation of MN technology in medicine.
Collapse
Affiliation(s)
- Fatma Moawad
- Faculté de Pharmacie, Université de Montréal, 2940 Chemin de Polytechnique, Montréal, Québec H3T 1J4, Canada; Faculty of Pharmacy, Beni-Suef University, Beni-Suef 625617, Egypt
| | - Roxane Pouliot
- Faculté de Pharmacie, Université Laval, Québec G1V 0A6, Canada
| | - Davide Brambilla
- Faculté de Pharmacie, Université de Montréal, 2940 Chemin de Polytechnique, Montréal, Québec H3T 1J4, Canada.
| |
Collapse
|
6
|
Beilmann M, Adkins K, Boonen HCM, Hewitt P, Hu W, Mader R, Moore S, Rana P, Steger-Hartmann T, Villenave R, van Vleet T. Application of new approach methodologies for nonclinical safety assessment of drug candidates. Nat Rev Drug Discov 2025:10.1038/s41573-025-01182-9. [PMID: 40316753 DOI: 10.1038/s41573-025-01182-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/12/2025] [Indexed: 05/04/2025]
Abstract
The development of new approach methodologies (NAMs) and advances with in vitro testing systems have prompted revisions in regulatory guidelines and inspired dedicated in vitro/ex vivo studies for nonclinical safety assessment. This Review by a safety reflection initiative subgroup of the European Federation of Pharmaceutical Industries and Associations (EFPIA)/Preclinical Development Expert Group (PDEG) summarizes the current state and potential application of in vitro studies using human-derived material for safety assessment in drug development. It focuses on case studies from recent projects in which animal models alone proved to be limited or inadequate for safety testing. It further highlights four categories of drug candidates for which alternative in vitro approaches are applicable and discusses progress in using in vitro testing solutions for safety assessment in these categories. Finally, the article highlights new risk assessment strategies, initiatives and consortia promoting the advancement of NAMs. This collective work is meant to encourage the use of NAMs for more human-relevant safety assessment, which should ultimately result in reduced animal testing for drug development.
Collapse
Affiliation(s)
- Mario Beilmann
- Global Nonclinical Safety & DMPK, Boehringer Ingelheim Pharma GmbH & Co. KG, Biberach an der Riss, Germany.
| | | | | | - Philip Hewitt
- Chemical and Preclinical Safety, Merck Healthcare KGaA, Darmstadt, Germany
| | - Wenyue Hu
- Vividion Therapeutics, San Diego, CA, USA
| | - Robert Mader
- Roche Pharma Research and Early Development, Roche Innovation Center Basel, F. Hoffmann-La Roche Ltd, Basel, Switzerland
| | | | - Payal Rana
- Drug Safety R&D, Pfizer Inc., Groton, CT, USA
| | - Thomas Steger-Hartmann
- Research & Development, Pharmaceuticals, Preclinical Development, Bayer AG, Berlin, Germany
| | - Remi Villenave
- Roche Pharma Research and Early Development, Roche Innovation Center Basel, F. Hoffmann-La Roche Ltd, Basel, Switzerland
| | | |
Collapse
|
7
|
Emborg ME, Metzger JM, D'Amour K, Colwell JC, Neumann LC, Zhang A, Federoff HJ. Advantages and challenges of using allogeneic vs. autologous sources for neuronal cell replacement in Parkinson's disease: Insights from non-human primate studies. Brain Res Bull 2025; 224:111297. [PMID: 40086764 PMCID: PMC12036832 DOI: 10.1016/j.brainresbull.2025.111297] [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: 09/25/2024] [Revised: 02/25/2025] [Accepted: 03/09/2025] [Indexed: 03/16/2025]
Abstract
Intracerebral grafting of dopamine-producing cells is proposed as a strategy to replace the typical neurons lost to Parkinson's disease (PD) and improve PD motor symptoms. Non-human primate studies have provided clues on the relationship between the host's immune response and grafting success. Herein, we discuss how the host's immune system differentially affects the graft depending on the origin of the cells and reflect on the advantages and limitations of the immune paradigms utilized to assess graft-related outcomes. We also consider new strategies to minimize or circumvent the host's immunological response and related preclinical research needed to identify the most promising new approaches to be translated into the clinic.
Collapse
Affiliation(s)
- Marina E Emborg
- Preclinical Parkinson's Research Program, Wisconsin National Primate Research Center, University of Wisconsin-Madison, USA; Department of Medical Physics, University of Wisconsin-Madison, USA; Cellular and Molecular Pathology Graduate Program, University of Wisconsin-Madison, USA.
| | - Jeanette M Metzger
- Preclinical Parkinson's Research Program, Wisconsin National Primate Research Center, University of Wisconsin-Madison, USA
| | | | - Julia C Colwell
- Preclinical Parkinson's Research Program, Wisconsin National Primate Research Center, University of Wisconsin-Madison, USA; Cellular and Molecular Pathology Graduate Program, University of Wisconsin-Madison, USA
| | - Lindsey C Neumann
- Preclinical Parkinson's Research Program, Wisconsin National Primate Research Center, University of Wisconsin-Madison, USA
| | - Ai Zhang
- Genentech, South San Francisco, CA, USA
| | - Howard J Federoff
- Kenai Therapeutics, San Diego, CA, USA; Georgetown University Medical Center, Washington, DC, USA
| |
Collapse
|
8
|
Hu X, Tediashvili G, Gravina A, Stoddard J, McGill TJ, Connolly AJ, Deuse T, Schrepfer S. Inhibition of polymorphonuclear cells averts cytotoxicity against hypoimmune cells in xenotransplantation. Nat Commun 2025; 16:3706. [PMID: 40251154 PMCID: PMC12008267 DOI: 10.1038/s41467-025-58774-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2024] [Accepted: 03/31/2025] [Indexed: 04/20/2025] Open
Abstract
Allogeneic, immune-evasive hypoimmune (HIP) cell therapeutics that are HLA-depleted and overexpress CD47 create the opportunity to treat immunocompetent patients with cancer, degenerative, or autoimmune diseases. However, HIP cell therapy has not yet been established for xenotransplantation. Here we engineer, for human-to-non-human primate studies, human HIP* endothelial cells (EC) that are HLA-depleted and express macaque CD47 to allow compatibility with the macaque SIRPα immune checkpoint. Although no T cell, NK cell, or macrophage responses and no antibody-dependent cytotoxicity is observed in cynomolgus recipients, we reveal that macaque polymorphonuclear cells (PMN) show strong xenogeneic cytotoxicity against HIP* ECs. Inhibition of PMN killing using a multi-drug regimen leads to improved xenogeneic human HIP* EC survival in cynomolgus monkeys. Similarly, human PMNs show xenoreactivity against pig ECs, which has implications for clinical xenotransplantation. Accordingly, our engineered pig HIP* ECs that are SLA-depleted, overexpress human CD47, and additionally overexpress the PMN-inhibitory ligands CD99 and CD200, are protected against all human adaptive and innate cytotoxicity, including PMNs. In summary, specific targeting of PMN-mediated killing of the transplanted cells might improve outcomes for clinical pig-to-human xenotransplantation.
Collapse
Affiliation(s)
- Xiaomeng Hu
- Department of Surgery, Division of Cardiothoracic Surgery, Transplant and Stem Cell Immunobiology (TSI)-Lab, University of California San Francisco, San Francisco, CA, USA
- Sana Biotechnology Inc., South San Francisco, CA, USA
| | - Grigol Tediashvili
- Department of Surgery, Division of Cardiothoracic Surgery, Transplant and Stem Cell Immunobiology (TSI)-Lab, University of California San Francisco, San Francisco, CA, USA
| | - Alessia Gravina
- Department of Surgery, Division of Cardiothoracic Surgery, Transplant and Stem Cell Immunobiology (TSI)-Lab, University of California San Francisco, San Francisco, CA, USA
| | - Jonathan Stoddard
- Division of Neuroscience, Oregon National Primate Research Center, Oregon Health & Science University, Portland, OR, USA
| | - Trevor J McGill
- Division of Neuroscience, Oregon National Primate Research Center, Oregon Health & Science University, Portland, OR, USA
| | - Andrew J Connolly
- Department of Pathology, University of California San Francisco, San Francisco, CA, USA
| | - Tobias Deuse
- Department of Surgery, Division of Cardiothoracic Surgery, Transplant and Stem Cell Immunobiology (TSI)-Lab, University of California San Francisco, San Francisco, CA, USA
| | - Sonja Schrepfer
- Department of Surgery, Division of Cardiothoracic Surgery, Transplant and Stem Cell Immunobiology (TSI)-Lab, University of California San Francisco, San Francisco, CA, USA.
- Sana Biotechnology Inc., South San Francisco, CA, USA.
- Department of Surgery, Cedars-Sinai Medical Center, Los Angeles, CA, USA.
- Board of Governors Regenerative Medicine Institute, Cedars-Sinai Medical Center, Los Angeles, CA, USA.
| |
Collapse
|
9
|
Gong JR, Lee J, Han Y, Cho KH. DDX54 downregulation enhances anti-PD1 therapy in immune-desert lung tumors with high tumor mutational burden. Proc Natl Acad Sci U S A 2025; 122:e2412310122. [PMID: 40172969 PMCID: PMC12002276 DOI: 10.1073/pnas.2412310122] [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: 06/19/2024] [Accepted: 02/25/2025] [Indexed: 04/04/2025] Open
Abstract
High tumor mutational burden (TMB-H) is a predictive biomarker for the responsiveness of cancer to immune checkpoint inhibitor (ICI) therapy that indicates whether immune cells can sufficiently recognize cancer cells as nonself. However, about 30% of all cancers from The Cancer Genome Atlas (TCGA) are classified as immune-desert tumors lacking T cell infiltration despite TMB-H. Since the underlying mechanism of these immune-desert tumors has yet to be unraveled, there is a pressing need to transform such immune-desert tumors into immune-inflamed tumors and thereby enhance their responsiveness to anti-PD1 therapy. Here, we present a systems framework for identifying immuno-oncotargets, based on analysis of gene regulatory networks, and validating the effect of these targets in transforming immune-desert into immune-inflamed tumors. In particular, we identify DEAD-box helicases 54 (DDX54) as a master regulator of immune escape in immune-desert lung cancer with TMB-H and show that knockdown of DDX54 can increase immune cell infiltration and lead to improved sensitivity to anti-PD1 therapy.
Collapse
Affiliation(s)
- Jeong-Ryeol Gong
- Department of Bio and Brain Engineering, Korea Advanced Institute of Science and Technology, Daejeon34141, Republic of Korea
| | - Jungeun Lee
- Department of Bio and Brain Engineering, Korea Advanced Institute of Science and Technology, Daejeon34141, Republic of Korea
| | - Younghyun Han
- Department of Bio and Brain Engineering, Korea Advanced Institute of Science and Technology, Daejeon34141, Republic of Korea
| | - Kwang-Hyun Cho
- Department of Bio and Brain Engineering, Korea Advanced Institute of Science and Technology, Daejeon34141, Republic of Korea
| |
Collapse
|
10
|
Yeapuri P, Machhi J, Foster EG, Kadry R, Bhattarai S, Lu Y, Sil S, Sapkota R, Srivastava S, Kumar M, Ikezu T, Poluektova LY, Gendelman HE, Mosley RL. Amyloid precursor protein and presenilin-1 knock-in immunodeficient mice exhibit intraneuronal Aβ pathology, microgliosis, and extensive neuronal loss. Alzheimers Dement 2025; 21:e70084. [PMID: 40195277 PMCID: PMC11975631 DOI: 10.1002/alz.70084] [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: 11/15/2024] [Revised: 02/13/2025] [Accepted: 02/17/2025] [Indexed: 04/09/2025]
Abstract
INTRODUCTION Transgenic mice overexpressing familial Alzheimer's disease (AD) mutations (FAD) show non-physiological traits, and their immunocompetent backgrounds limit their use in AD immunotherapy research. Preclinical models that reflect human immune responses in AD are needed. METHODS Using CRISPR-Cas9, we developed single (NA) and double (NAPS) knock-in (KI) amyloid precursor protein (APP)KM670,671NL (Swedish) and presenilin 1 (PS 1)M146VFAD mutations on an immunodeficient NOG (NOD.Cg-PrkdcscidIl2rgtm1Sug/JicTac) background. The models were confirmed by Sanger sequencing and evaluated for AD-like pathology. RESULTS Both NA and NAPS mice developed pathology without overexpression artifacts. Mutation-induced upregulation of APP-CTF-β led to intraneuronal human amyloid beta (Aβ) (6E10) deposits and amyloid-associated microgliosis as early as 3 months, which increased with age. The addition of the PS 1M146V mutation doubled the amyloid load. The models displayed broad neuronal loss, resulting in brain atrophy in older mice. DISCUSSION These models replicate intraneuronal amyloid pathology and, with human immune reconstitution potential, enable novel studies of human immune responses in AD. HIGHLIGHTS A novel Alzheimer's disease (AD) knock-in (KI) mouse was developed and characterized on an immunodeficient NOG background. The model provides a platform for human immune studies and the evaluation of immunotherapies for AD. The KI mice demonstrate intraneuronal Aβ deposits and amyloid-associated microglial reactions. KI mice demonstrate extensive neuronal loss. Human immune reconstitution enables studies of infectious AD co-morbidities, such as the human immunodeficiency and herpes simplex viruses.
Collapse
Affiliation(s)
- Pravin Yeapuri
- Department of Pharmacology and Experimental NeuroscienceCenter for Neurodegenerative DisordersCollege of MedicineUniversity of Nebraska Medical CenterOmahaNebraskaUSA
| | - Jatin Machhi
- Department of Pharmacology and Experimental NeuroscienceCenter for Neurodegenerative DisordersCollege of MedicineUniversity of Nebraska Medical CenterOmahaNebraskaUSA
| | - Emma G. Foster
- Department of Pharmacology and Experimental NeuroscienceCenter for Neurodegenerative DisordersCollege of MedicineUniversity of Nebraska Medical CenterOmahaNebraskaUSA
| | - Rana Kadry
- Department of Pharmacology and Experimental NeuroscienceCenter for Neurodegenerative DisordersCollege of MedicineUniversity of Nebraska Medical CenterOmahaNebraskaUSA
| | - Shaurav Bhattarai
- Department of Pharmacology and Experimental NeuroscienceCenter for Neurodegenerative DisordersCollege of MedicineUniversity of Nebraska Medical CenterOmahaNebraskaUSA
| | - Yaman Lu
- Department of Pharmacology and Experimental NeuroscienceCenter for Neurodegenerative DisordersCollege of MedicineUniversity of Nebraska Medical CenterOmahaNebraskaUSA
| | - Susmita Sil
- Department of Pharmacology and Experimental NeuroscienceCenter for Neurodegenerative DisordersCollege of MedicineUniversity of Nebraska Medical CenterOmahaNebraskaUSA
| | - Roshan Sapkota
- Department of Pharmacology and Experimental NeuroscienceCenter for Neurodegenerative DisordersCollege of MedicineUniversity of Nebraska Medical CenterOmahaNebraskaUSA
| | - Shefali Srivastava
- Department of Pharmacology and Experimental NeuroscienceCenter for Neurodegenerative DisordersCollege of MedicineUniversity of Nebraska Medical CenterOmahaNebraskaUSA
| | - Mohit Kumar
- Department of Pharmacology and Experimental NeuroscienceCenter for Neurodegenerative DisordersCollege of MedicineUniversity of Nebraska Medical CenterOmahaNebraskaUSA
| | - Tsuneya Ikezu
- Department of NeuroscienceMayo Clinic FloridaJacksonvilleFloridaUSA
| | - Larisa Y. Poluektova
- Department of Pharmacology and Experimental NeuroscienceCenter for Neurodegenerative DisordersCollege of MedicineUniversity of Nebraska Medical CenterOmahaNebraskaUSA
| | - Howard E. Gendelman
- Department of Pharmacology and Experimental NeuroscienceCenter for Neurodegenerative DisordersCollege of MedicineUniversity of Nebraska Medical CenterOmahaNebraskaUSA
| | - Rodney Lee Mosley
- Department of Pharmacology and Experimental NeuroscienceCenter for Neurodegenerative DisordersCollege of MedicineUniversity of Nebraska Medical CenterOmahaNebraskaUSA
| |
Collapse
|
11
|
Intonti S, Kokona D, Zinkernagel MS, Enzmann V, Stein JV, Conedera FM. Glia Modulates Immune Responses in the Retina Through Distinct MHC Pathways. Glia 2025; 73:822-839. [PMID: 39873321 PMCID: PMC11845847 DOI: 10.1002/glia.24656] [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: 08/07/2024] [Revised: 11/22/2024] [Accepted: 11/25/2024] [Indexed: 01/30/2025]
Abstract
Glia antigen-presenting cells (APCs) are pivotal regulators of immune surveillance within the retina, maintaining tissue homeostasis and promptly responding to insults. However, the intricate mechanisms underlying their local coordination and activation remain unclear. Our study integrates an animal model of retinal injury, retrospective analysis of human retinas, and in vitro experiments to gain insights into the crucial role of antigen presentation in neuroimmunology during retinal degeneration (RD), uncovering the involvement of various glial cells, notably Müller glia and microglia. Glial cells act as sentinels, detecting antigens released during degeneration and interacting with T-cells via MHC molecules, which are essential for immune responses. Microglia function as APCs via the MHC Class II pathway, upregulating key molecules such as Csf1r and cytokines. In contrast, Müller cells act through the MHC Class I pathway, exhibiting upregulated antigen processing genes and promoting a CD8+ T-cell response. Distinct cytokine signaling pathways, including TNF-α and IFN Type I, contribute to the immune balance. Human retinal specimens corroborate these findings, demonstrating glial activation and MHC expression correlating with degenerative changes. In vitro assays also confirmed differential T-cell migration responses to activated microglia and Müller cells, highlighting their role in shaping the immune milieu within the retina. In summary, our study emphasizes the involvement of retinal glial cells in modulating the immune response after insults to the retinal parenchyma. Unraveling the intricacies of glia-mediated antigen presentation in RD is essential for developing precise therapeutic interventions for retinal pathologies.
Collapse
Affiliation(s)
- Simona Intonti
- Department of Experimental and Clinical MedicineUniversity of FlorenceFlorenceItaly
- Department of Ophthalmology, Bern University Hospital and Department of BioMedical ResearchUniversity of BernBernSwitzerland
| | - Despina Kokona
- Department of Ophthalmology, Bern University Hospital and Department of BioMedical ResearchUniversity of BernBernSwitzerland
| | - Martin S. Zinkernagel
- Department of Ophthalmology, Bern University Hospital and Department of BioMedical ResearchUniversity of BernBernSwitzerland
| | - Volker Enzmann
- Department of Ophthalmology, Bern University Hospital and Department of BioMedical ResearchUniversity of BernBernSwitzerland
| | - Jens V. Stein
- Department of Oncology, Microbiology and ImmunologyUniversity of FribourgFribourgSwitzerland
| | - Federica M. Conedera
- Department of Ophthalmology, Bern University Hospital and Department of BioMedical ResearchUniversity of BernBernSwitzerland
- Department of Oncology, Microbiology and ImmunologyUniversity of FribourgFribourgSwitzerland
| |
Collapse
|
12
|
Papenfuss TL, Moghe AP, Himmel LE, Goyos A, Weinstock D. Immunotoxicology From a Pathology Perspective: A Continuing Education Course Presented at the Annual STP Meeting in Baltimore, MD, Held June 16-19, 2024. Toxicol Pathol 2025:1926233251328970. [PMID: 40159901 DOI: 10.1177/01926233251328970] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/02/2025]
Abstract
This article is a summary of a half-day continuing education course jointly sponsored by the Society for Toxicologic Pathology (STP) and the Health and Environmental Sciences Institute (HESI) at the annual meeting of the STP in Baltimore, MD, held June 16-19, 2024. Presenters discussed pathology, toxicology, immunotoxicology, and regulatory implications of findings in the immune system in context of development of immunomodulatory therapeutics. Interpretation of pathology findings requires knowledge of immune system morphology and function including species-specific differences and spontaneous findings in animal model systems. A weight of evidence (WoE) approach is required to integrate pathology findings and immunotoxicology assay results to assess translatability to humans. Communication and collaboration among scientists of various disciplines can be instrumental in optimal generation and interpretation of appropriate data for development of immunomodulatory therapeutics.
Collapse
Affiliation(s)
| | | | | | - Ana Goyos
- Johnson and Johnson Innovative Medicine, La Jolla, California, USA
| | | |
Collapse
|
13
|
Meier RPH, Ben Nasr M, Fife BT, Finger EB, Fiorina P, Luo X, Bromberg JS. Best practices in islet transplantation in mice. Am J Transplant 2025:S1600-6135(25)00137-6. [PMID: 40089068 DOI: 10.1016/j.ajt.2025.03.008] [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: 10/20/2024] [Revised: 01/30/2025] [Accepted: 03/07/2025] [Indexed: 03/17/2025]
Abstract
Islet transplantation in mice serves as a crucial preclinical model for understanding alloimmune and autoimmune mechanisms, optimizing immunosuppressive strategies, and developing novel therapies for diabetes. This review provides a comprehensive overview of best practices in murine islet transplantation, including diabetes induction models, technical aspects of islet transplantation, and criteria for transplant graft and rejection. We discuss the immunological challenges posed by major histocompatibility complex disparities, the impact of various transplantation sites, and the limitations of murine models in translating findings to clinical settings. Special emphasis is placed on emerging strategies such as stem cell-derived insulin-producing cells, immune tolerance induction, and alternative transplantation sites. Although mouse models have significantly advanced our understanding of diabetes and β-cell replacement, their inherent differences from human physiology necessitate careful interpretation of findings. The review also highlights novel imaging modalities, immunosuppressive protocols, and biomarkers for graft monitoring, underscoring the need for further refinement of these models to bridge the gap between experimental research and clinical application. By standardizing methodologies and addressing translational limitations, murine islet transplantation studies remain a key model in transplantation and can continue to shape the future of β-cell replacement therapies for insulin-dependent diabetes.
Collapse
Affiliation(s)
- Raphael P H Meier
- Department of Surgery, University of Maryland, School of Medicine, Baltimore, Maryland, USA.
| | - Moufida Ben Nasr
- International Center for T1D, Pediatric Clinical Research Center Romeo ed Enrica Invernizzi, DIBIC, Università di Milano, Milan, Italy; Nephrology Division, Boston Children's Hospital, Harvard Medical School, Boston, Massachusetts, USA; Transplantation Research Center, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Brian T Fife
- Department of Medicine, Division of Rheumatic and Autoimmune Diseases, University of Minnesota Medical School, Minneapolis, Minnesota, USA; Center for Immunology, University of Minnesota Medical School, Minneapolis, Minnesota, USA
| | - Erik B Finger
- Department of Surgery, University of Minnesota, Minneapolis, Minnesota, USA
| | - Paolo Fiorina
- International Center for T1D, Pediatric Clinical Research Center Romeo ed Enrica Invernizzi, DIBIC, Università di Milano, Milan, Italy; Nephrology Division, Boston Children's Hospital, Harvard Medical School, Boston, Massachusetts, USA; Transplantation Research Center, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Xunrong Luo
- Department of Medicine, Duke University, Durham, North Carolina, USA
| | - Jonathan S Bromberg
- Department of Surgery, University of Maryland, School of Medicine, Baltimore, Maryland, USA.
| |
Collapse
|
14
|
Guan F, Wang R, Yi Z, Luo P, Liu W, Xie Y, Liu Z, Xia Z, Zhang H, Cheng Q. Tissue macrophages: origin, heterogenity, biological functions, diseases and therapeutic targets. Signal Transduct Target Ther 2025; 10:93. [PMID: 40055311 PMCID: PMC11889221 DOI: 10.1038/s41392-025-02124-y] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2024] [Revised: 11/01/2024] [Accepted: 12/15/2024] [Indexed: 05/04/2025] Open
Abstract
Macrophages are immune cells belonging to the mononuclear phagocyte system. They play crucial roles in immune defense, surveillance, and homeostasis. This review systematically discusses the types of hematopoietic progenitors that give rise to macrophages, including primitive hematopoietic progenitors, erythro-myeloid progenitors, and hematopoietic stem cells. These progenitors have distinct genetic backgrounds and developmental processes. Accordingly, macrophages exhibit complex and diverse functions in the body, including phagocytosis and clearance of cellular debris, antigen presentation, and immune response, regulation of inflammation and cytokine production, tissue remodeling and repair, and multi-level regulatory signaling pathways/crosstalk involved in homeostasis and physiology. Besides, tumor-associated macrophages are a key component of the TME, exhibiting both anti-tumor and pro-tumor properties. Furthermore, the functional status of macrophages is closely linked to the development of various diseases, including cancer, autoimmune disorders, cardiovascular disease, neurodegenerative diseases, metabolic conditions, and trauma. Targeting macrophages has emerged as a promising therapeutic strategy in these contexts. Clinical trials of macrophage-based targeted drugs, macrophage-based immunotherapies, and nanoparticle-based therapy were comprehensively summarized. Potential challenges and future directions in targeting macrophages have also been discussed. Overall, our review highlights the significance of this versatile immune cell in human health and disease, which is expected to inform future research and clinical practice.
Collapse
Affiliation(s)
- Fan Guan
- Department of Neurosurgery, Xiangya Hospital, Central South University, Changsha, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China
- Xiangya School of Medicine, Central South University, Changsha, China
| | - Ruixuan Wang
- Department of Neurosurgery, Xiangya Hospital, Central South University, Changsha, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China
| | - Zhenjie Yi
- Department of Neurosurgery, Xiangya Hospital, Central South University, Changsha, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China
| | - Peng Luo
- Department of Oncology, Zhujiang Hospital, Southern Medical University, Guangzhou, China
| | - Wanyao Liu
- Xiangya School of Medicine, Central South University, Changsha, China
| | - Yao Xie
- Xiangya School of Medicine, Central South University, Changsha, China
| | - Zaoqu Liu
- Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Zhiwei Xia
- Department of Neurology, Hunan Aerospace Hospital, Hunan Normal University, Changsha, China.
| | - Hao Zhang
- Department of Neurosurgery, The Second Affiliated Hospital, Chongqing Medical University, Chongqing, China.
| | - Quan Cheng
- Department of Neurosurgery, Xiangya Hospital, Central South University, Changsha, China.
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China.
| |
Collapse
|
15
|
Nisar MF, Yan T, Cai Y, Wan C. Immuno-oncological Challenges and Chemoresistance in Veterinary Medicine: Probiotics as a New Strategic Tool. Probiotics Antimicrob Proteins 2025:10.1007/s12602-025-10468-8. [PMID: 39954194 DOI: 10.1007/s12602-025-10468-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/21/2025] [Indexed: 02/17/2025]
Abstract
Cancer has the highest death rates due to increased immuno-oncological (IO) challenges and chemoresistance caused by gut dysbiosis, whereas administration of probiotics may reverse these responses against anticancer therapies. Recently, immunotherapeutics have extensively been focused for significant advancements in pharmacological drug discovery and clinical outcomes. Mammals have intestinal epithelial cells, mucosal immune cells, and indigenous gut microbiota which may reshape immunotherapeutics efficacy. These include use of T-cell immune checkpoint inhibitors (ICPI), genetically engineered T-cells, tumor vaccines, monoclonal antibodies (mAbs), and anti-B- and T-cell antibodies. Immunotherapeutics for cancer treatment became popular in both veterinary and human health care systems due to their strong inhibitory actions against PD-1 and CTLA-4 to check tumorigenesis. IO issues in animals also need special attention, where caninized mAbs targeting CD-20 and CD-52 have been clinically used in treating canine B-cell and T-cell lymphomas, respectively. Probiotics appeared as strong immunotherapeutics that might be shaping the epigenetics of the organisms specifically in animal breeding practices for desired features, but limited literature regarding the immunomodulatory effects in humans and animals is available. In addition, considering the important role of probiotics in humans and veterinary medicine, a new perspective on the probiotic-mediated modulation of ncRNAs (miRNAs, lncRNAs, circRNAs) is also highlighted and would be a new therapeutic tool. This review provides insight into the cellular processes and pharmacological activities for treating veterinary infectious diseases and covers small drug molecules as ncRNA-modulators in veterinary medicine.
Collapse
Affiliation(s)
- Muhammad Farrukh Nisar
- Ministry of Education and Jiangxi Key Laboratory of Crop Physiology, Ecology and Genetic Breeding, Jiangxi Agricultural University, Nanchang, 330045, China
- Jiangxi Key Laboratory for Post-harvest Technology and Nondestructive Testing of Fruits & Vegetables, College of Agronomy, Jiangxi Agricultural University, Nanchang, 330045, China
- Department of Physiology and Biochemistry, Cholistan University of Veterinary and Animal Sciences (CUVAS), Bahawalpur, Pakistan
| | - Tingdong Yan
- School of Pharmacy, Nantong University, Nantong, 226001, China.
| | - Yi Cai
- Guangzhou Municipal and Guangdong Provincial Key Laboratory of Molecular Target & Clinical Pharmacology, the NMPA and State Key Laboratory of Respiratory Disease, School of Pharmaceutical Sciences and the Fifth Affiliated Hospital, Guangzhou Medical University, Guangzhou, 511436, China
| | - Chunpeng Wan
- Jiangxi Key Laboratory for Post-harvest Technology and Nondestructive Testing of Fruits & Vegetables, College of Agronomy, Jiangxi Agricultural University, Nanchang, 330045, China.
| |
Collapse
|
16
|
Aksoy SA, Earl J, Grahovac J, Karakas D, Lencioni G, Sığırlı S, Bijlsma MF. Organoids, tissue slices and organotypic cultures: Advancing our understanding of pancreatic ductal adenocarcinoma through in vitro and ex vivo models. Semin Cancer Biol 2025; 109:10-24. [PMID: 39730107 DOI: 10.1016/j.semcancer.2024.12.003] [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: 10/04/2024] [Revised: 12/14/2024] [Accepted: 12/19/2024] [Indexed: 12/29/2024]
Abstract
Pancreatic ductal adenocarcinoma (PDAC) has one of the worst prognoses of all common solid cancers. For the large majority of PDAC patients, only systemic therapies with very limited efficacy are indicated. In addition, immunotherapies have not brought the advances seen in other cancer types. Several key characteristics of PDAC contribute to poor treatment outcomes, and in this review, we will discuss how these characteristics are best captured in currently available ex vivo or in vitro model systems. For instance, PDAC is hallmarked by a highly desmoplastic and immune-suppressed tumor microenvironment that impacts disease progression and therapy resistance. Also, large differences in tumor biology exist between and within tumors, complicating treatment decisions. Furthermore, PDAC has a very high propensity for locally invasive and metastatic growth. The use of animal models is often not desirable or feasible and several in vitro and ex vivo model systems have been developed, such as organotypic cocultures and tissue slices, among others. However, the absence of a full host organism impacts the ability of these models to accurately capture the characteristics that contribute to poor outcomes in PDAC. We will discuss the caveats and advantages of these model systems in the context of PDAC's key characteristics and provide recommendations on model choice and the possibilities for optimization. These considerations should be of use to researchers aiming to study PDAC in the in vitro setting.
Collapse
Affiliation(s)
- Secil Ak Aksoy
- Bursa Uludag University, Faculty of Medicine, Department of Medical Microbiology, Bursa, Turkey
| | - Julie Earl
- Ramón y Cajal Health Research Institute (IRYCIS), Biomodels and Biomodels Platform Hospital Ramón y Cajal-IRYCIS, Carretera Colmenar Km 9,100, Madrid 28034, Spain; The Biomedical Research Network in Cancer (CIBERONC), Av. Monforte de Lemos, 3-5. Pabellón 11. Planta 0, Madrid 28029, Spain
| | - Jelena Grahovac
- Experimental Oncology Department, Institute for Oncology and Radiology of Serbia, Belgrade, Serbia
| | - Didem Karakas
- Acibadem Mehmet Ali Aydinlar University, Department of Medical Biotechnology, Graduate School of Health Sciences, Istanbul, Turkey
| | - Giulia Lencioni
- Department of Biology, University of Pisa, Pisa, Italy; Fondazione Pisana per la Scienza, San Giuliano Terme, Pisa, Italy
| | - Sıla Sığırlı
- Acibadem Mehmet Ali Aydinlar University, Department of Medical Biotechnology, Graduate School of Health Sciences, Istanbul, Turkey
| | - Maarten F Bijlsma
- Amsterdam UMC Location University of Amsterdam, Laboratory of Experimental Oncology and Radiobiology, Amsterdam, the Netherlands; Cancer Center Amsterdam, Cancer Biology, Amsterdam, the Netherlands.
| |
Collapse
|
17
|
Katz LN, Bohlen MO, Yu G, Mejias-Aponte C, Sommer MA, Krauzlis RJ. Optogenetic Manipulation of Covert Attention in the Nonhuman Primate. J Cogn Neurosci 2025; 37:266-285. [PMID: 39509098 PMCID: PMC12022921 DOI: 10.1162/jocn_a_02274] [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: 11/15/2024]
Abstract
Optogenetics affords new opportunities to interrogate neuronal circuits that control behavior. In primates, the usefulness of optogenetics in studying cognitive functions remains a challenge. The technique has been successfully wielded, but behavioral effects have been demonstrated primarily for sensorimotor processes. Here, we tested whether brief optogenetic suppression of primate superior colliculus can change performance in a covert attention task, in addition to previously reported optogenetic effects on saccadic eye movements. We used an attention task that required the monkey to detect and report a stimulus change at a cued location via joystick release, while ignoring changes at an uncued location. When the cued location was positioned in the response fields of transduced neurons in the superior colliculus, transient light delivery coincident with the stimulus change disrupted the monkey's detection performance, significantly lowering hit rates. When the cued location was elsewhere, hit rates were unaltered, indicating that the effect was spatially specific and not a motor deficit. Hit rates for trials with only one stimulus were also unaltered, indicating that the effect depended on selection among distractors rather than a low-level visual impairment. Psychophysical analysis revealed that optogenetic suppression increased perceptual threshold, but only for locations matching the transduced site. These data show that optogenetic manipulations can cause brief and spatially specific deficits in covert attention, independent of sensorimotor functions. This dissociation of effect, and the temporal precision provided by the technique, demonstrates the utility of optogenetics in interrogating neuronal circuits that mediate cognitive functions in the primate.
Collapse
|
18
|
Naumova DA, Krokunova T, Maksimov D, Mityaeva ON, Astakhova EA, Volchkov PY. Challenges in Humoral Immune Response to Adeno-Associated Viruses Determination. Int J Mol Sci 2025; 26:816. [PMID: 39859531 PMCID: PMC11765838 DOI: 10.3390/ijms26020816] [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: 12/06/2024] [Revised: 01/10/2025] [Accepted: 01/17/2025] [Indexed: 01/27/2025] Open
Abstract
Adeno-associated viruses (AAVs) are non-pathogenic, replication-deficient viruses that have gained widespread attention for their application as gene therapy vectors. While these vectors offer high transduction efficiency and long-term gene expression, the host immune response poses a significant challenge to their clinical success. This review focuses on the obstacles to evaluating the humoral response to AAVs. We discuss the problems with the validation of in vitro tests and the possible approaches to overcome them. Using published data on neutralizing titers of AAV serotypes, we built the first antigenic maps of AAVs in order to visualize the antigenic relationships between varying serotypes.
Collapse
Affiliation(s)
- Daria A. Naumova
- Federal Research Center for Original and Prospective Biomedical and Pharmaceutical Technologies, 8 Baltiyskaya Street, Moscow 125315, Russia
- Moscow Center for Advanced Studies, Kulakova Street, 20, Moscow 123592, Russia
| | - Tatyana Krokunova
- Federal Research Center for Original and Prospective Biomedical and Pharmaceutical Technologies, 8 Baltiyskaya Street, Moscow 125315, Russia
- Moscow Center for Advanced Studies, Kulakova Street, 20, Moscow 123592, Russia
| | - Denis Maksimov
- Federal Research Center for Original and Prospective Biomedical and Pharmaceutical Technologies, 8 Baltiyskaya Street, Moscow 125315, Russia
- Moscow Center for Advanced Studies, Kulakova Street, 20, Moscow 123592, Russia
| | - Olga N. Mityaeva
- Federal Research Center for Original and Prospective Biomedical and Pharmaceutical Technologies, 8 Baltiyskaya Street, Moscow 125315, Russia
- Moscow Center for Advanced Studies, Kulakova Street, 20, Moscow 123592, Russia
| | - Ekaterina A. Astakhova
- Federal Research Center for Original and Prospective Biomedical and Pharmaceutical Technologies, 8 Baltiyskaya Street, Moscow 125315, Russia
- Moscow Center for Advanced Studies, Kulakova Street, 20, Moscow 123592, Russia
| | - Pavel Yu Volchkov
- Federal Research Center for Original and Prospective Biomedical and Pharmaceutical Technologies, 8 Baltiyskaya Street, Moscow 125315, Russia
- Moscow Center for Advanced Studies, Kulakova Street, 20, Moscow 123592, Russia
| |
Collapse
|
19
|
Anisimov AP, Vagaiskaya AS, Trunyakova AS, Dentovskaya SV. Live Plague Vaccine Development: Past, Present, and Future. Vaccines (Basel) 2025; 13:66. [PMID: 39852845 PMCID: PMC11768842 DOI: 10.3390/vaccines13010066] [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: 12/02/2024] [Revised: 01/04/2025] [Accepted: 01/08/2025] [Indexed: 01/26/2025] Open
Abstract
During the last 100 years, vaccine development has evolved from an empirical approach to one of the more rational vaccine designs where the careful selection of antigens and adjuvants is key to the desired efficacy for challenging pathogens and/or challenging populations. To improve immunogenicity while maintaining a favorable reactogenicity and safety profile, modern vaccine design must consider factors beyond the choice of target antigen alone. With new vaccine technologies currently emerging, it will be possible to custom-design vaccines for optimal efficacy in groups of people with different responses to vaccination. It should be noted that after a fairly long period of overwhelming dominance of papers devoted to subunit plague vaccines, materials devoted to the development of live plague vaccines have increasingly been published. In this review, we present our opinion on reasonable tactics for the development and application of live, safe, and protective human plague vaccines causing an enhanced duration of protection and breadth of action against various virulent strains in vaccination studies representing different ages, genders, and nucleotide polymorphisms of the genes responsible for immune response.
Collapse
Affiliation(s)
- Andrey P. Anisimov
- Laboratory for Plague Microbiology, Especially Dangerous Infections Department, State Research Center for Applied Microbiology and Biotechnology, 142279 Obolensk, Russia; (A.S.V.); (A.S.T.); (S.V.D.)
| | | | | | | |
Collapse
|
20
|
Mulholland MM, Nehete BP, DeLise A, Achorn AM, Pytka LM, Nehete PN. Age-associated alterations in immune and inflammatory responses in captive olive baboons ( Papio anubis). FRONTIERS IN AGING 2025; 5:1511370. [PMID: 39835299 PMCID: PMC11743478 DOI: 10.3389/fragi.2024.1511370] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 10/14/2024] [Accepted: 12/16/2024] [Indexed: 01/22/2025]
Abstract
Introduction Advanced age is a primary risk factor for many chronic diseases and conditions; however, age-related immune dysregulation is not well understood. Animal models, particularly those that resemble human age-related physiological changes, are needed to better understand immunosenescence and to improve health outcomes. Here, we explore the utility of the olive baboon (Papio anubis) in studying age-related changes to the immune system and understanding mechanisms of immunosenescence. Methods We examined immune cell, inflammatory responses, cytokines, and cortisol levels using hematology and flow cytometry, mitogen stimulation, multiplex cytokine assay, and cortisol immunoassay. Results and Discussion Our results reveal significant age effects on numerous immune and inflammatory responses. For instance, adult and aged monkeys exhibited significantly fewer monocytes than young monkeys. After stimulation with Con A and PWM (separately), we found that old baboons had higher INFγ expression compared to young baboons. Similarly, after stimulation with LPS and PWM (separately), we found that old baboons had higher TNFα expression compared to young baboons. These findings suggest that the olive baboon is a suitable model for biogerontology research, immune senescence, and development of vaccines. Though there are phenotypic and functional similarities between baboons and humans, specific differences exist in immune cell expression and immune function of lymphocytes that should be considered for better experimental outcomes in the development of therapeutics and restoring innate and adaptive immune function in aged individuals.
Collapse
Affiliation(s)
- Michele M. Mulholland
- The University of Texas MD Anderson Cancer Center Bastrop, Department of Comparative Medicine TX, Bastrop, TX, United States
| | - Bharti P. Nehete
- The University of Texas MD Anderson Cancer Center Bastrop, Department of Comparative Medicine TX, Bastrop, TX, United States
| | - Ashley DeLise
- The University of Texas MD Anderson Cancer Center Bastrop, Department of Comparative Medicine TX, Bastrop, TX, United States
| | - Angela M. Achorn
- The University of Texas MD Anderson Cancer Center Bastrop, Department of Comparative Medicine TX, Bastrop, TX, United States
| | - Lisa M. Pytka
- The University of Texas MD Anderson Cancer Center Bastrop, Department of Comparative Medicine TX, Bastrop, TX, United States
| | - Pramod N. Nehete
- The University of Texas MD Anderson Cancer Center Bastrop, Department of Comparative Medicine TX, Bastrop, TX, United States
- The University of Texas Graduate School of Biomedical Sciences at Houston, Houston, TX, United States
| |
Collapse
|
21
|
Youhanna S, Kemas AM, Wright SC, Zhong Y, Klumpp B, Klein K, Motso A, Michel M, Ziegler N, Shang M, Sabatier P, Kannt A, Sheng H, Oliva‐Vilarnau N, Büttner FA, Seashore‐Ludlow B, Schreiner J, Windbergs M, Cornillet M, Björkström NK, Hülsmeier AJ, Hornemann T, Olsen JV, Wang Y, Gramignoli R, Sundström M, Lauschke VM. Chemogenomic Screening in a Patient-Derived 3D Fatty Liver Disease Model Reveals the CHRM1-TRPM8 Axis as a Novel Module for Targeted Intervention. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2025; 12:e2407572. [PMID: 39605182 PMCID: PMC11744578 DOI: 10.1002/advs.202407572] [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: 07/05/2024] [Revised: 09/19/2024] [Indexed: 11/29/2024]
Abstract
Metabolic dysfunction-associated steatohepatitis (MASH) is a leading cause of chronic liver disease with few therapeutic options. To narrow the translational gap in the development of pharmacological MASH treatments, a 3D liver model from primary human hepatocytes and non-parenchymal cells derived from patients with histologically confirmed MASH was established. The model closely mirrors disease-relevant endpoints, such as steatosis, inflammation and fibrosis, and multi-omics analyses show excellent alignment with biopsy data from 306 MASH patients and 77 controls. By combining high-content imaging with scalable biochemical assays and chemogenomic screening, multiple novel targets with anti-steatotic, anti-inflammatory, and anti-fibrotic effects are identified. Among these, activation of the muscarinic M1 receptor (CHRM1) and inhibition of the TRPM8 cation channel result in strong anti-fibrotic effects, which are confirmed using orthogonal genetic assays. Strikingly, using biosensors based on bioluminescence resonance energy transfer, a functional interaction along a novel MASH signaling axis in which CHRM1 inhibits TRPM8 via Gq/11 and phospholipase C-mediated depletion of phosphatidylinositol 4,5-bisphosphate can be demonstrated. Combined, this study presents the first patient-derived 3D MASH model, identifies a novel signaling module with anti-fibrotic effects, and highlights the potential of organotypic culture systems for phenotype-based chemogenomic drug target identification at scale.
Collapse
|
22
|
Yu S, Xie J, Li PH, Chen Y, Tang IY, Lin X. Therapeutic potential of interleukin-17 neutralization in a novel humanized mouse model of Sjögren's disease. Pharmacol Res 2024; 210:107524. [PMID: 39617280 DOI: 10.1016/j.phrs.2024.107524] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/16/2024] [Revised: 11/26/2024] [Accepted: 11/26/2024] [Indexed: 12/06/2024]
Abstract
Sjögren's disease (SjD) is a chronic autoimmune disease, in which the immune system targets exocrine glands and leads to dryness symptoms. There is an increasing need to develop novel therapeutic approach as the treatment plan has not been changed in the past decade. However, findings in mouse model may not be directly applied in patients, given the substantial differences of immune system between human and mice. In the present study, using antigens derived from human salivary A-253 cells, we established experimental Sjögren's syndrome (ESS) in mice with human immune system (HIS). HIS-ESS mice exhibited key features of human disease, including salivary hypofunction, increased serum levels of autoantibodies and tissue destruction in the salivary glands. Phenotypic analysis revealed enhanced effector B and T cell subsets, including Th1, Th17 and T follicular helper (Tfh) cells in HIS-ESS mice, while multiplex imaging analysis suggested enlarged B cell follicles and expanded memory B cells. IL-17 neutralization therapy significantly ameliorated disease pathology at both acute and chronic stages, in which B cells were mainly affected, to the less extent Th1 and Tfh cells in HIS-ESS mice. Together, HIS-ESS mouse model highly recapitulated SjD features and immunopathogenesis, which may serve as a useful tool in drug screening and pre-clinical studies.
Collapse
Affiliation(s)
- Sulan Yu
- School of Chinese Medicine, the University of Hong Kong, Hong Kong
| | - Jing Xie
- School of Chinese Medicine, the University of Hong Kong, Hong Kong
| | - Philip Hei Li
- Division of Rheumatology and Clinical Immunology, Department of Medicine, Queen Mary Hospital, The University of Hong Kong, Hong Kong
| | - Yacun Chen
- School of Chinese Medicine, the University of Hong Kong, Hong Kong
| | - Iris Yanki Tang
- School of Chinese Medicine, the University of Hong Kong, Hong Kong
| | - Xiang Lin
- School of Chinese Medicine, the University of Hong Kong, Hong Kong; State Key Laboratory of Pharmaceutical Biotechnology, The University of Hong Kong, Hong Kong.
| |
Collapse
|
23
|
Salvador-Martínez I, Murga-Moreno J, Nieto JC, Alsinet C, Enard D, Heyn H. Adaptation in human immune cells residing in tissues at the frontline of infections. Nat Commun 2024; 15:10329. [PMID: 39609395 PMCID: PMC11605006 DOI: 10.1038/s41467-024-54603-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2023] [Accepted: 11/14/2024] [Indexed: 11/30/2024] Open
Abstract
Human immune cells are under constant evolutionary pressure, primarily through their role as first line of defence against pathogens. Most studies on immune adaptation are, however, based on protein-coding genes without considering their cellular context. Here, using data from the Human Cell Atlas, we infer the gene adaptation rate of the human immune landscape at cellular resolution. We find abundant cell types, like progenitor cells during development and adult cells in barrier tissues, to harbour significantly increased adaptation rates. We confirm the adaptation of tissue-resident T and NK cells in the adult lung located in compartments directly facing external challenges, such as respiratory pathogens. Analysing human iPSC-derived macrophages responding to various challenges, we find adaptation in early immune responses. Together, our study suggests host benefits to control pathogen spread at early stages of infection, providing a retrospect of forces that shaped the complexity, architecture, and function of the human body.
Collapse
Affiliation(s)
| | - Jesus Murga-Moreno
- Department of Ecology and Evolutionary Biology, University of Arizona, Tucson, AZ, USA
| | - Juan C Nieto
- CNAG, Centro Nacional de Análisis Genómico, Barcelona, Spain
| | - Clara Alsinet
- CNAG, Centro Nacional de Análisis Genómico, Barcelona, Spain
| | - David Enard
- Department of Ecology and Evolutionary Biology, University of Arizona, Tucson, AZ, USA.
| | - Holger Heyn
- CNAG, Centro Nacional de Análisis Genómico, Barcelona, Spain.
- Universitat de Barcelona (UB), Barcelona, Spain.
- ICREA, Barcelona, Spain.
| |
Collapse
|
24
|
Peralta-Álvarez MP, Downward K, White A, Redondo Azema H, Sibley L, Sarfas C, Morrison A, Dennis M, Diaz-Santana D, Harris SA, Li S, Puentes E, Aguilo N, Martin C, Sharpe S, McShane H, Tanner R. MTBVAC induces superior antibody titers and IgG avidity compared to BCG vaccination in non-human primates. NPJ Vaccines 2024; 9:230. [PMID: 39567530 PMCID: PMC11579480 DOI: 10.1038/s41541-024-01009-5] [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: 07/04/2024] [Accepted: 10/22/2024] [Indexed: 11/22/2024] Open
Abstract
The only currently licensed vaccine against tuberculosis (TB), Bacille Calmette Guérin (BCG), is insufficient to control the epidemic. MTBVAC is a live attenuated strain of Mycobacterium tuberculosis (M.tb) and is one the most advanced TB vaccine candidates in the pipeline. It is more efficacious than BCG in preclinical models including non-human primates (NHPs), and has demonstrated safety and immunogenicity in human populations. To better understand the immune mechanisms underlying the superior efficacy conferred by MTBVAC, we characterized M.tb-specific antibody responses in NHPs vaccinated with either BCG or MTBVAC. MTBVAC vaccination induced higher titers of IgG, IgM and IgA, and higher avidity IgG compared with BCG vaccination. IgG avidity correlated with protection following M.tb challenge in the same animals, validating the association previously reported between this measure and protection in the context of intravenous BCG vaccination, suggesting that IgG avidity may represent a relevant marker or correlate of protection from TB.
Collapse
Affiliation(s)
- Marco Polo Peralta-Álvarez
- Jenner Institute, Nuffield Department of Medicine, University of Oxford, Oxford, UK
- Laboratorio Nacional de Vacunologia y Virus Tropicales, Departamento de Microbiologia, Escuela Nacional de Ciencias Biologicas, Instituto Politecnico Nacional, Ciudad de Mexico, Mexico
| | - Keya Downward
- Jenner Institute, Nuffield Department of Medicine, University of Oxford, Oxford, UK
| | - Andrew White
- UK Health Security Agency, Porton Down, Salisbury, UK
| | - Hugo Redondo Azema
- Jenner Institute, Nuffield Department of Medicine, University of Oxford, Oxford, UK
- École Normale Supérieure - PSL, Paris, France
| | - Laura Sibley
- UK Health Security Agency, Porton Down, Salisbury, UK
| | | | | | - Mike Dennis
- UK Health Security Agency, Porton Down, Salisbury, UK
| | | | - Stephanie A Harris
- Jenner Institute, Nuffield Department of Medicine, University of Oxford, Oxford, UK
| | - Shuailin Li
- Jenner Institute, Nuffield Department of Medicine, University of Oxford, Oxford, UK
| | - Eugenia Puentes
- Clinical Research Department y Research and Development Department, Biofabri, Grupo Zendal, O'Porriño, Pontevedra, Spain
| | - Nacho Aguilo
- Faculty of Medicine, University of Zaragoza, Zaragoza, CIBERES, Instituto de Salud Carlos III, Madrid, Spain
| | - Carlos Martin
- Faculty of Medicine, University of Zaragoza, Zaragoza, CIBERES, Instituto de Salud Carlos III, Madrid, Spain
| | - Sally Sharpe
- UK Health Security Agency, Porton Down, Salisbury, UK
| | - Helen McShane
- Jenner Institute, Nuffield Department of Medicine, University of Oxford, Oxford, UK
| | - Rachel Tanner
- Jenner Institute, Nuffield Department of Medicine, University of Oxford, Oxford, UK.
- Department of Biology, University of Oxford, Oxford, UK.
| |
Collapse
|
25
|
Tu Q, Liu G, Liu X, Zhang J, Xiao W, Lv L, Zhao B. Perspective on using non-human primates in Exposome research. ECOTOXICOLOGY AND ENVIRONMENTAL SAFETY 2024; 286:117199. [PMID: 39426107 DOI: 10.1016/j.ecoenv.2024.117199] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/18/2024] [Revised: 08/02/2024] [Accepted: 10/13/2024] [Indexed: 10/21/2024]
Abstract
The physiological and pathological changes in the human body caused by environmental pressures are collectively referred to as the Exposome. Human society is facing escalating environmental pollution, leading to a rising prevalence of associated diseases, including respiratory diseases, cardiovascular diseases, neurological disorders, reproductive development disorders, among others. Vulnerable populations to the pathogenic effects of environmental pollution include those in the prenatal, infancy, and elderly stages of life. Conducting Exposome mechanistic research and proposing effective health interventions are urgent in addressing the current severe environmental pollution. In this review, we address the core issues and bottlenecks faced by current Exposome research, specifically focusing on the most toxic ultrafine nanoparticles. We summarize multiple research models being used in Exposome research. Especially, we discuss the limitations of rodent animal models in mimicking human physiopathological phenotypes, and prospect advantages and necessity of non-human primates in Exposome research based on their evolutionary relatedness, anatomical and physiological similarities to human. Finally, we declare the initiation of NHPE (Non-Human Primate Exposome) project for conducting Exposome research using non-human primates and provide insights into its feasibility and key areas of focus. SYNOPSIS: Non-human primate models hold unique advantages in human Exposome research.
Collapse
Affiliation(s)
- Qiu Tu
- Key Laboratory of Genetic Evolution & Animal Models, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, Yunnan 650223, China; Key Laboratory of Animal Models and Human Disease Mechanisms of the Chinese Academy of Sciences & Yunnan Province, and KIZ-CUHK Joint Laboratory of Bioresources and Molecular Research in Common Diseases, Kunming, Yunnan 650223, China
| | - Gaojing Liu
- Key Laboratory of Genetic Evolution & Animal Models, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, Yunnan 650223, China; Key Laboratory of Animal Models and Human Disease Mechanisms of the Chinese Academy of Sciences & Yunnan Province, and KIZ-CUHK Joint Laboratory of Bioresources and Molecular Research in Common Diseases, Kunming, Yunnan 650223, China; University of Chinese Academy of Sciences, Beijing 101408, China
| | - Xiuyun Liu
- Key Laboratory of Genetic Evolution & Animal Models, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, Yunnan 650223, China; Key Laboratory of Animal Models and Human Disease Mechanisms of the Chinese Academy of Sciences & Yunnan Province, and KIZ-CUHK Joint Laboratory of Bioresources and Molecular Research in Common Diseases, Kunming, Yunnan 650223, China; University of Chinese Academy of Sciences, Beijing 101408, China
| | - Jiao Zhang
- Key Laboratory of Genetic Evolution & Animal Models, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, Yunnan 650223, China; Key Laboratory of Animal Models and Human Disease Mechanisms of the Chinese Academy of Sciences & Yunnan Province, and KIZ-CUHK Joint Laboratory of Bioresources and Molecular Research in Common Diseases, Kunming, Yunnan 650223, China
| | - Wenxian Xiao
- Key Laboratory of Genetic Evolution & Animal Models, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, Yunnan 650223, China; Primate Facility, National Research Facility for Phenotypic & Genetic Analysis of Model Animals, and National Resource Center for Non-Human Primates, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, Yunnan 650223, China; National Resource Center for Non-Human Primates, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, Yunnan 650107, China
| | - Longbao Lv
- Key Laboratory of Genetic Evolution & Animal Models, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, Yunnan 650223, China; Primate Facility, National Research Facility for Phenotypic & Genetic Analysis of Model Animals, and National Resource Center for Non-Human Primates, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, Yunnan 650223, China; National Resource Center for Non-Human Primates, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, Yunnan 650107, China.
| | - Bo Zhao
- Key Laboratory of Genetic Evolution & Animal Models, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, Yunnan 650223, China; Key Laboratory of Animal Models and Human Disease Mechanisms of the Chinese Academy of Sciences & Yunnan Province, and KIZ-CUHK Joint Laboratory of Bioresources and Molecular Research in Common Diseases, Kunming, Yunnan 650223, China; Primate Facility, National Research Facility for Phenotypic & Genetic Analysis of Model Animals, and National Resource Center for Non-Human Primates, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, Yunnan 650223, China.
| |
Collapse
|
26
|
Paudyal B, Moorhouse E, Sharma B, Dodds M, Nguyen V, Milad M, Tchilian E. Comparative pharmacokinetics of porcine and human anti-influenza hemagglutinin monoclonal antibodies in outbred pigs and minipigs. Front Immunol 2024; 15:1471412. [PMID: 39544926 PMCID: PMC11560753 DOI: 10.3389/fimmu.2024.1471412] [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: 07/27/2024] [Accepted: 09/30/2024] [Indexed: 11/17/2024] Open
Abstract
Assessing the pharmacokinetics of monoclonal antibodies (mAbs) in relevant animal models is essential for designing improved formulations and developing mAb delivery platforms. We have established the pig, a large natural host animal for influenza with many similarities to humans, as a robust model for testing the therapeutic efficacy of anti-influenza mAbs and evaluating mAb delivery platforms. Here, we compared the pharmacokinetic characteristics of two anti-influenza hemagglutinin mAbs, human 2-12C and porcine pb27, in Göttingen minipigs and Landrace × Large White outbred pigs. Minipigs offer the advantage of a more stable weight, whereas outbred pigs are more readily available but exhibit rapid growth. Outbred pigs and minipigs showed similar pharmacokinetics and a similar porcine pb27 half-life (half-life of 15.7 days for outbred pigs and 16.6 days for minipigs). In contrast, the half-life of human 2-12C was more rapid in two of the minipigs but not in the outbred pigs, correlating with the development of antidrug antibodies in the two minipigs. Our results demonstrate that both outbred pigs and minipigs are appropriate models for pharmacokinetic studies and the evaluation of mAb delivery platforms, potentially bridging the gap between small animals and human trials.
Collapse
Affiliation(s)
- Basudev Paudyal
- Host Responses, The Pirbright Institute, Woking, United Kingdom
| | | | - Bhawna Sharma
- Host Responses, The Pirbright Institute, Woking, United Kingdom
| | - Michael Dodds
- Integrated Drug Development, Certara, Radnor, PA, United States
| | - Victor Nguyen
- Milad Pharmaceutical Consulting LLC, Plymouth, MI, United States
| | - Mark Milad
- Milad Pharmaceutical Consulting LLC, Plymouth, MI, United States
| | - Elma Tchilian
- Host Responses, The Pirbright Institute, Woking, United Kingdom
| |
Collapse
|
27
|
Rivero Berti I, Gambaro RC, Limeres MJ, Huck-Iriart C, Svensson M, Fraude-El Ghazi S, Pretsch L, Si S, Lieberwirth I, Landfester K, Cacicedo ML, Islan GA, Gehring S. Encapsulation of Dexamethasone into mRNA-Lipid Nanoparticles Is a Promising Approach for the Development of Liver-Targeted Anti-Inflammatory Therapies. Int J Mol Sci 2024; 25:11254. [PMID: 39457035 PMCID: PMC11508592 DOI: 10.3390/ijms252011254] [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: 09/23/2024] [Revised: 10/06/2024] [Accepted: 10/17/2024] [Indexed: 10/28/2024] Open
Abstract
The objective of this study was to develop two lipid nanoparticle (LNP) formulations capable of efficiently expressing a reporter mRNA while co-delivering the anti-inflammatory drug dexamethasone (DX) to reduce inflammatory side effects in protein replacement therapies. Two types of LNPs were developed, in which 25% of cholesterol was replaced by DX. These LNPs contained either 1,2-distearoyl-sn-glycero-3-phosphocholine (DSPC) or 1,2-dioleoyl-sn-glycero-3-phosphoethanolamine (DOPE) as a helper lipid. The resulting LNPs exhibited high stability, homogeneity, and near-neutral Zeta potentials. SAXS experiments confirmed DX incorporation into the LNP core, with slow in vitro DX release observed over 48 h. The LNPs achieved high mRNA encapsulation efficiency (95-100%) and effectively transfected HepG2 cells, dendritic cells, and hPBMCs. While LNPs increased cytokine release (IL-1β, TNF-α, MCP-1), LNPs-DX significantly reduced cytokine levels, demonstrating enhanced anti-inflammatory properties while maintaining mRNA expression levels. In vivo biodistribution showed predominant liver localization post-intramuscular injection, regardless of the DSPC or DOPE composition. LNPs co-loaded with mRNA and DX are promising candidates for continuous protein replacement. Due to their ability to reduce treatment-related inflammation while maintaining significant mRNA expression levels, these LNPs are perfectly suited for the treatment of liver-related metabolic diseases.
Collapse
Affiliation(s)
- Ignacio Rivero Berti
- Children’s Hospital, University Medical Center of the Johannes Gutenberg University, Langenbeckstr. 1, 55131 Mainz, Germany; (I.R.B.); (R.C.G.); (M.J.L.); (M.S.); (S.F.-E.G.); (L.P.); (M.L.C.)
- CINDEFI—Centro de Investigación y Desarrollo en Fermentaciones Industriales, Laboratorio de Nanobiomateriales, Facultad de Ciencias Exactas, Universidad Nacional de La Plata (UNLP)-Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), La Plata 1900, Argentina
| | - Rocío Celeste Gambaro
- Children’s Hospital, University Medical Center of the Johannes Gutenberg University, Langenbeckstr. 1, 55131 Mainz, Germany; (I.R.B.); (R.C.G.); (M.J.L.); (M.S.); (S.F.-E.G.); (L.P.); (M.L.C.)
| | - María José Limeres
- Children’s Hospital, University Medical Center of the Johannes Gutenberg University, Langenbeckstr. 1, 55131 Mainz, Germany; (I.R.B.); (R.C.G.); (M.J.L.); (M.S.); (S.F.-E.G.); (L.P.); (M.L.C.)
| | - Cristián Huck-Iriart
- ALBA Synchrotron Light Source, Carrer de la Llum 2–26, Cerdanyola del Vallès, 08290 Barcelona, Spain;
| | - Malin Svensson
- Children’s Hospital, University Medical Center of the Johannes Gutenberg University, Langenbeckstr. 1, 55131 Mainz, Germany; (I.R.B.); (R.C.G.); (M.J.L.); (M.S.); (S.F.-E.G.); (L.P.); (M.L.C.)
| | - Silvia Fraude-El Ghazi
- Children’s Hospital, University Medical Center of the Johannes Gutenberg University, Langenbeckstr. 1, 55131 Mainz, Germany; (I.R.B.); (R.C.G.); (M.J.L.); (M.S.); (S.F.-E.G.); (L.P.); (M.L.C.)
| | - Leah Pretsch
- Children’s Hospital, University Medical Center of the Johannes Gutenberg University, Langenbeckstr. 1, 55131 Mainz, Germany; (I.R.B.); (R.C.G.); (M.J.L.); (M.S.); (S.F.-E.G.); (L.P.); (M.L.C.)
| | - Shutian Si
- Max Planck Institute for Polymer Research, Department of Physical Chemistry of Polymers, Ackermannweg 10, 55128 Mainz, Germany; (S.S.); (I.L.); (K.L.)
| | - Ingo Lieberwirth
- Max Planck Institute for Polymer Research, Department of Physical Chemistry of Polymers, Ackermannweg 10, 55128 Mainz, Germany; (S.S.); (I.L.); (K.L.)
| | - Katharina Landfester
- Max Planck Institute for Polymer Research, Department of Physical Chemistry of Polymers, Ackermannweg 10, 55128 Mainz, Germany; (S.S.); (I.L.); (K.L.)
| | - Maximiliano Luis Cacicedo
- Children’s Hospital, University Medical Center of the Johannes Gutenberg University, Langenbeckstr. 1, 55131 Mainz, Germany; (I.R.B.); (R.C.G.); (M.J.L.); (M.S.); (S.F.-E.G.); (L.P.); (M.L.C.)
| | - Germán Abel Islan
- Children’s Hospital, University Medical Center of the Johannes Gutenberg University, Langenbeckstr. 1, 55131 Mainz, Germany; (I.R.B.); (R.C.G.); (M.J.L.); (M.S.); (S.F.-E.G.); (L.P.); (M.L.C.)
- CINDEFI—Centro de Investigación y Desarrollo en Fermentaciones Industriales, Laboratorio de Nanobiomateriales, Facultad de Ciencias Exactas, Universidad Nacional de La Plata (UNLP)-Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), La Plata 1900, Argentina
| | - Stephan Gehring
- Children’s Hospital, University Medical Center of the Johannes Gutenberg University, Langenbeckstr. 1, 55131 Mainz, Germany; (I.R.B.); (R.C.G.); (M.J.L.); (M.S.); (S.F.-E.G.); (L.P.); (M.L.C.)
| |
Collapse
|
28
|
Alb M, Reiche K, Rade M, Sewald K, Loskill P, Cipriano M, Maulana TI, van der Meer AD, Weener HJ, Clerbaux LA, Fogal B, Patel N, Adkins K, Lund E, Perkins E, Cooper C, van den Brulle J, Morgan H, Rubic-Schneider T, Ling H, DiPetrillo K, Moggs J, Köhl U, Hudecek M. Novel strategies to assess cytokine release mediated by chimeric antigen receptor T cells based on the adverse outcome pathway concept. J Immunotoxicol 2024; 21:S13-S28. [PMID: 39655500 DOI: 10.1080/1547691x.2024.2345158] [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: 07/10/2023] [Revised: 02/21/2024] [Accepted: 04/15/2024] [Indexed: 12/18/2024] Open
Abstract
The success of cellular immunotherapies such as chimeric antigen receptor (CAR) T cell therapy has led to their implementation as a revolutionary treatment option for cancer patients. However, the safe translation of such novel immunotherapies, from non-clinical assessment to first-in-human studies is still hampered by the lack of suitable in vitro and in vivo models recapitulating the complexity of the human immune system. Additionally, using cells derived from human healthy volunteers in such test systems may not adequately reflect the altered state of the patient's immune system thus potentially underestimating the risk of life-threatening conditions, such as cytokine release syndrome (CRS) following CAR T cell therapy. The IMI2/EU project imSAVAR (immune safety avatar: non-clinical mimicking of the immune system effects of immunomodulatory therapies) aims at creating a platform for novel tools and models for enhanced non-clinical prediction of possible adverse events associated with immunomodulatory therapies. This platform shall in the future guide early non-clinical safety assessment of novel immune therapeutics thereby also reducing the costs of their development. Therefore, we review current opportunities and challenges associated with non-clinical in vitro and in vivo models for the safety assessment of CAR T cell therapy ranging from organ-on-chip models up to advanced biomarker screening.
Collapse
MESH Headings
- Humans
- Receptors, Chimeric Antigen/immunology
- Receptors, Chimeric Antigen/genetics
- Immunotherapy, Adoptive/methods
- Immunotherapy, Adoptive/adverse effects
- Cytokine Release Syndrome/immunology
- Cytokine Release Syndrome/therapy
- Cytokine Release Syndrome/diagnosis
- Animals
- T-Lymphocytes/immunology
- Neoplasms/therapy
- Neoplasms/immunology
- Cytokines/metabolism
- Cytokines/immunology
- Receptors, Antigen, T-Cell/immunology
- Receptors, Antigen, T-Cell/metabolism
- Receptors, Antigen, T-Cell/genetics
Collapse
Affiliation(s)
- Miriam Alb
- Medizinische Klinik und Poliklinik II, Lehrstuhl für Zelluläre Immuntherapie, Universitätsklinikum Würzburg, Würzburg, Germany
| | - Kristin Reiche
- Fraunhofer-Institut für Zelltherapie und Immunologie IZI, Leipzig, Germany
| | - Michael Rade
- Fraunhofer-Institut für Zelltherapie und Immunologie IZI, Leipzig, Germany
| | - Katherina Sewald
- Fraunhofer-Institut für Toxikologie und Experimentelle Medizin ITEM, Hannover, Germany
| | - Peter Loskill
- Institute for Biomedical Engineering, Eberhard Karls University Tübingen, Tübingen, Germany
- 3R-Center for In vitro Models and Alternatives to Animal Testing, Eberhard Karls University Tübingen, Tübingen
- NMI Natural and Medical Sciences Institute at the University of Tübingen, Reutlingen, Germany
| | - Madalena Cipriano
- Institute for Biomedical Engineering, Eberhard Karls University Tübingen, Tübingen, Germany
- 3R-Center for In vitro Models and Alternatives to Animal Testing, Eberhard Karls University Tübingen, Tübingen
| | - Tengku Ibrahim Maulana
- Institute for Biomedical Engineering, Eberhard Karls University Tübingen, Tübingen, Germany
- 3R-Center for In vitro Models and Alternatives to Animal Testing, Eberhard Karls University Tübingen, Tübingen
| | | | - Huub J Weener
- Applied Stem Cell Technologies, University of Twente, Enschede, the Netherlands
| | | | - Birgit Fogal
- Department on Nonclinical Drug Safety, Boehringer Ingelheim Pharmaceutical, Inc, Ridgefield, CT, USA
| | - Nirav Patel
- Preclinical Safety, Research and Development, Sanofi-Aventis US, LLC, Cambridge, MA, USA
| | - Karissa Adkins
- Preclinical Safety, Research and Development, Sanofi-Aventis US, LLC, Cambridge, MA, USA
| | - Emma Lund
- Labcorp Drug Development Inc, Derbyshire, UK
| | | | | | | | - Hannah Morgan
- Novartis Biomedical Research, Novartis Campus, Basel, Switzerland
| | | | - Hui Ling
- Novartis Biomedical Research, Cambridge, MA, USA
| | | | - Jonathan Moggs
- Novartis Biomedical Research, Novartis Campus, Basel, Switzerland
| | - Ulrike Köhl
- Fraunhofer-Institut für Zelltherapie und Immunologie IZI, Leipzig, Germany
| | - Michael Hudecek
- Medizinische Klinik und Poliklinik II, Lehrstuhl für Zelluläre Immuntherapie, Universitätsklinikum Würzburg, Würzburg, Germany
- Fraunhofer-Institut für Zelltherapie und Immunologie IZI, Leipzig, Germany
| |
Collapse
|
29
|
Creppy JR, Delache B, Lemaitre J, Horvat B, Vecellio L, Ducancel F. Administration of airborne pathogens in non-human primates. Inhal Toxicol 2024; 36:475-500. [PMID: 39388247 DOI: 10.1080/08958378.2024.2412685] [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] [Accepted: 09/30/2024] [Indexed: 10/15/2024]
Abstract
PURPOSE Airborne pathogen scan penetrate in human respiratory tract and can cause illness. The use of animal models to predict aerosol deposition and study respiratory disease pathophysiology is therefore important for research and a prerequisite to test and study the mechanism of action of treatment. NHPs are relevant animal species for inhalation studies because of their similarities with humans in terms of anatomical structure, respiratory parameters and immune system. MATERIALS AND METHODS The aim of this review is to provide an overview of the state of the art of pathogen aerosol studies performed in non-human primates (NHPs). Herein, we present and discuss the deposition of aerosolized bacteria and viruses. In this review, we present important advantages of using NHPs as model for inhalation studies. RESULTS We demonstrate that deposition in the respiratory tract is not only a function of aerosol size but also the technique of administration influences the biological activity and site of aerosol deposition. Finally, we observe an influence of a region of pathogen deposition in the respiratory tract on the development of the pathophysiological effect in NHPs. CONCLUSION The wide range of methods used for the delivery of pathogento NHP respiratory airways is associated with varying doses and deposition profiles in the airways.
Collapse
Affiliation(s)
- Justina R Creppy
- Inserm, CEA, Center for Immunology of Viral, Auto-immune, Hematological and Bacterial Diseases (IMVA-HB/IDMIT), Université Paris-Saclay, Fontenay-aux-Roses, France
- Centre d'Étude des Pathologies Respiratoires, INSERM U1100, Université de Tours, Tours, France
| | - Benoit Delache
- Inserm, CEA, Center for Immunology of Viral, Auto-immune, Hematological and Bacterial Diseases (IMVA-HB/IDMIT), Université Paris-Saclay, Fontenay-aux-Roses, France
| | - Julien Lemaitre
- Inserm, CEA, Center for Immunology of Viral, Auto-immune, Hematological and Bacterial Diseases (IMVA-HB/IDMIT), Université Paris-Saclay, Fontenay-aux-Roses, France
| | - Branka Horvat
- CIRI, Centre International de Recherche en Infectiologie, INSERM U1111, CNRS UMR5308, Université de Lyon, Lyon, France
| | - Laurent Vecellio
- Centre d'Étude des Pathologies Respiratoires, INSERM U1100, Université de Tours, Tours, France
| | - Frédéric Ducancel
- Inserm, CEA, Center for Immunology of Viral, Auto-immune, Hematological and Bacterial Diseases (IMVA-HB/IDMIT), Université Paris-Saclay, Fontenay-aux-Roses, France
| |
Collapse
|
30
|
Girkin CA, Strickland RG, Somerville MM, Garner MA, Grossman GH, Blake A, Kumar N, Ianov L, Fazio MA, Clark ME, Gross AK. Acute ocular hypertension in the living human eye: Model description and initial cellular responses to elevated intraocular pressure. Vision Res 2024; 223:108465. [PMID: 39173459 PMCID: PMC11444249 DOI: 10.1016/j.visres.2024.108465] [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: 06/05/2024] [Revised: 08/02/2024] [Accepted: 08/02/2024] [Indexed: 08/24/2024]
Abstract
This initial methods study presents the initial immunohistochemical and transcriptomic changes in the optic nerve head and retina from three research-consented brain-dead organ donors following prolonged and transient intraocular pressure (IOP) elevation. In this initial study, research-consented brain-dead organ donors were exposed to unilateral elevation of IOP for 7.5 h (Donor 1), 30 h (Donor 2), and 1 h (Donor 3) prior to organ procurement. Optic nerve tissue and retinal tissue was obtained following organ procurement for immunohistological and transcriptomic analysis. Optic nerve sections in Donor 1 exposed to 7.5-hours of unilateral sub-ischemic IOP elevation demonstrated higher levels of protein expression of the astrocytic marker, glial fibrillary acidic protein (GFAP), within the lamina cribrosa with greatest expression inferior temporally in the treated eye compared to control. Spatial transcriptomic analysis performed on optic nerve head tissues from Donor 2 exposed to 30 h of unilateral IOP elevation demonstrated differential transcription of mRNA across laminar and scleral regions. Immunohistochemistry of retinal sections from Donor 2 exhibited higher GFAP and IBA1 expression in the treated eye compared with control, but this was not observed in Donor 3, which was exposed to only 1-hour of IOP elevation. While there were no differences in GFAP protein expression in the retina following the 1-hour IOP elevation in Donor 3, there were higher levels of transcription of GFAP in the inner nuclear layer, and CD44 in the retinal ganglion cell layer, indicative of astrocytic and Müller glial reactivity as well as an early inflammatory response, respectively. We found that transcriptomic differences can be observed across treated and control eyes following unilateral elevation of IOP in brain dead organ donors. The continued development of this model affords the unique opportunity to define the acute mechanotranscriptomic response of the optic nerve head, evaluate the injury and repair mechanisms in the retina in response to IOP elevation, and enable correlation of in vivo imaging and functional testing with ex vivo cellular responses for the first time in the living human eye.
Collapse
Affiliation(s)
- Christopher A Girkin
- Department of Ophthalmology and Visual Sciences, Heersink School of Medicine, University of Alabama at Birmingham, Birmingham, AL, USA; Department of Neurobiology, Heersink School of Medicine, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Ryan G Strickland
- Department of Neurobiology, Heersink School of Medicine, University of Alabama at Birmingham, Birmingham, AL, USA
| | - McKenna M Somerville
- Department of Neurobiology, Heersink School of Medicine, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Mary Anne Garner
- Department of Ophthalmology and Visual Sciences, Heersink School of Medicine, University of Alabama at Birmingham, Birmingham, AL, USA; Department of Neurobiology, Heersink School of Medicine, University of Alabama at Birmingham, Birmingham, AL, USA
| | | | - Alan Blake
- Advancing Sight Network, Birmingham, AL, USA
| | - Nilesh Kumar
- IRCP-Biological Data Sciences, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Lara Ianov
- Department of Neurobiology, Heersink School of Medicine, University of Alabama at Birmingham, Birmingham, AL, USA; IRCP-Biological Data Sciences, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Massimo A Fazio
- Department of Ophthalmology and Visual Sciences, Heersink School of Medicine, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Mark E Clark
- Department of Ophthalmology and Visual Sciences, Heersink School of Medicine, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Alecia K Gross
- Department of Ophthalmology and Visual Sciences, Heersink School of Medicine, University of Alabama at Birmingham, Birmingham, AL, USA; Department of Neurobiology, Heersink School of Medicine, University of Alabama at Birmingham, Birmingham, AL, USA.
| |
Collapse
|
31
|
Stepanova E, Isakova-Sivak I, Matyushenko V, Mezhenskaya D, Kudryavtsev I, Kostromitina A, Chistiakova A, Rak A, Bazhenova E, Prokopenko P, Kotomina T, Donina S, Novitskaya V, Sivak K, Karal-Ogly D, Rudenko L. Safety and Immunogenicity Study of a Bivalent Vaccine for Combined Prophylaxis of COVID-19 and Influenza in Non-Human Primates. Vaccines (Basel) 2024; 12:1099. [PMID: 39460266 PMCID: PMC11511058 DOI: 10.3390/vaccines12101099] [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: 08/19/2024] [Revised: 09/23/2024] [Accepted: 09/24/2024] [Indexed: 10/28/2024] Open
Abstract
BACKGROUND Influenza and SARS-CoV-2 viruses are two highly variable pathogens. We have developed a candidate bivalent live vaccine based on the strain of licensed A/Leningrad/17-based cold-adapted live attenuated influenza vaccine (LAIV) of H3N2 subtype, which expressed SARS-CoV-2 immunogenic T-cell epitopes. A cassette encoding fragments of S and N proteins of SARS-CoV-2 was inserted into the influenza NA gene using the P2A autocleavage site. In this study, we present the results of preclinical evaluation of the developed bivalent vaccine in a non-human primate model. METHODS Rhesus macaques (Macaca mulatta) (n = 3 per group) were immunized intranasally with 7.5 lg EID50 of the LAIV/CoV-2 bivalent vaccine, a control non-modified H3N2 LAIV or a placebo (chorioallantoic fluid) using a sprayer device, twice, with a 28-day interval. The blood samples were collected at days 0, 3, 28 and 35 for hematological and biochemical assessment. Safety was also assessed by monitoring body weight, body temperature and clinical signs of the disease. Immune responses to influenza virus were assessed both by determining serum antibody titers in hemagglutination inhibition assay, microneutralization assay and IgG ELISA. T-cell responses were measured both to influenza and SARS-CoV-2 antigens using ELISPOT and flow cytometry. Three weeks after the second immunization, animals were challenged with 105 PFU of Delta SARS-CoV-2. The body temperature, weight and challenge virus shedding were monitored for 5 days post-challenge. In addition, virus titers in various organs and histopathology were evaluated on day 6 after SARS-CoV-2 infection. RESULTS There was no toxic effect of the immunizations on the hematological and coagulation hemostasis of animals. No difference in the dynamics of the average weight and thermometry results were found between the groups of animals. Both LAIV and LAIV/CoV-2 variants poorly replicated in the upper respiratory tract of rhesus macaques. Nevertheless, despite this low level of virus shedding, influenza-specific serum IgG responses were detected in the group of monkeys immunized with the LAIV/CoV-2 bivalent but not in the LAIV group. Furthermore, T-cell responses to both influenza and SARS-CoV-2 viruses were detected in the LAIV/CoV-2 vaccine group only. The animals were generally resistant to SARS-CoV-2 challenge, with minimal virus shedding in the placebo and LAIV groups. Histopathological changes in vaccinated animals were decreased compared to the PBS group, suggesting a protective effect of the chimeric vaccine candidate. CONCLUSIONS The candidate bivalent vaccine was safe and immunogenic for non-human primates and warrants its further evaluation in clinical trials.
Collapse
Affiliation(s)
- Ekaterina Stepanova
- Institute of Experimental Medicine, Saint-Petersburg 197022, Russia; (I.I.-S.); (V.M.); (D.M.); (I.K.); (A.K.); (A.C.); (A.R.); (P.P.); (T.K.); (V.N.); (L.R.)
| | - Irina Isakova-Sivak
- Institute of Experimental Medicine, Saint-Petersburg 197022, Russia; (I.I.-S.); (V.M.); (D.M.); (I.K.); (A.K.); (A.C.); (A.R.); (P.P.); (T.K.); (V.N.); (L.R.)
| | - Victoria Matyushenko
- Institute of Experimental Medicine, Saint-Petersburg 197022, Russia; (I.I.-S.); (V.M.); (D.M.); (I.K.); (A.K.); (A.C.); (A.R.); (P.P.); (T.K.); (V.N.); (L.R.)
| | - Daria Mezhenskaya
- Institute of Experimental Medicine, Saint-Petersburg 197022, Russia; (I.I.-S.); (V.M.); (D.M.); (I.K.); (A.K.); (A.C.); (A.R.); (P.P.); (T.K.); (V.N.); (L.R.)
| | - Igor Kudryavtsev
- Institute of Experimental Medicine, Saint-Petersburg 197022, Russia; (I.I.-S.); (V.M.); (D.M.); (I.K.); (A.K.); (A.C.); (A.R.); (P.P.); (T.K.); (V.N.); (L.R.)
| | - Arina Kostromitina
- Institute of Experimental Medicine, Saint-Petersburg 197022, Russia; (I.I.-S.); (V.M.); (D.M.); (I.K.); (A.K.); (A.C.); (A.R.); (P.P.); (T.K.); (V.N.); (L.R.)
| | - Anna Chistiakova
- Institute of Experimental Medicine, Saint-Petersburg 197022, Russia; (I.I.-S.); (V.M.); (D.M.); (I.K.); (A.K.); (A.C.); (A.R.); (P.P.); (T.K.); (V.N.); (L.R.)
| | - Alexandra Rak
- Institute of Experimental Medicine, Saint-Petersburg 197022, Russia; (I.I.-S.); (V.M.); (D.M.); (I.K.); (A.K.); (A.C.); (A.R.); (P.P.); (T.K.); (V.N.); (L.R.)
| | - Ekaterina Bazhenova
- Institute of Experimental Medicine, Saint-Petersburg 197022, Russia; (I.I.-S.); (V.M.); (D.M.); (I.K.); (A.K.); (A.C.); (A.R.); (P.P.); (T.K.); (V.N.); (L.R.)
| | - Polina Prokopenko
- Institute of Experimental Medicine, Saint-Petersburg 197022, Russia; (I.I.-S.); (V.M.); (D.M.); (I.K.); (A.K.); (A.C.); (A.R.); (P.P.); (T.K.); (V.N.); (L.R.)
| | - Tatiana Kotomina
- Institute of Experimental Medicine, Saint-Petersburg 197022, Russia; (I.I.-S.); (V.M.); (D.M.); (I.K.); (A.K.); (A.C.); (A.R.); (P.P.); (T.K.); (V.N.); (L.R.)
| | - Svetlana Donina
- Institute of Experimental Medicine, Saint-Petersburg 197022, Russia; (I.I.-S.); (V.M.); (D.M.); (I.K.); (A.K.); (A.C.); (A.R.); (P.P.); (T.K.); (V.N.); (L.R.)
| | - Vlada Novitskaya
- Institute of Experimental Medicine, Saint-Petersburg 197022, Russia; (I.I.-S.); (V.M.); (D.M.); (I.K.); (A.K.); (A.C.); (A.R.); (P.P.); (T.K.); (V.N.); (L.R.)
| | - Konstantin Sivak
- Smorodintsev Research Institute of Influenza, Saint-Petersburg 197376, Russia;
| | - Dzhina Karal-Ogly
- Center of Preclinical Research, Research Institute of Medical Primatology, Sochi 354376, Russia;
| | - Larisa Rudenko
- Institute of Experimental Medicine, Saint-Petersburg 197022, Russia; (I.I.-S.); (V.M.); (D.M.); (I.K.); (A.K.); (A.C.); (A.R.); (P.P.); (T.K.); (V.N.); (L.R.)
| |
Collapse
|
32
|
Evangelisti C, Ramadan S, Orlacchio A, Panza E. Experimental Cell Models for Investigating Neurodegenerative Diseases. Int J Mol Sci 2024; 25:9747. [PMID: 39273694 PMCID: PMC11396244 DOI: 10.3390/ijms25179747] [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: 08/16/2024] [Revised: 09/02/2024] [Accepted: 09/04/2024] [Indexed: 09/15/2024] Open
Abstract
Experimental models play a pivotal role in biomedical research, facilitating the understanding of disease mechanisms and the development of novel therapeutics. This is particularly true for neurodegenerative diseases, such as Alzheimer's disease, Parkinson's disease, Huntington's disease, amyotrophic lateral sclerosis, and motor neuron disease, which present complex challenges for research and therapy development. In this work, we review the recent literature about experimental models and motor neuron disease. We identified three main categories of models that are highly studied by scientists. In fact, experimental models for investigating these diseases encompass a variety of approaches, including modeling the patient's cell culture, patient-derived induced pluripotent stem cells, and organoids. Each model offers unique advantages and limitations, providing researchers with a range of tools to address complex biological questions. Here, we discuss the characteristics, applications, and recent advancements in terms of each model system, highlighting their contributions to advancing biomedical knowledge and translational research.
Collapse
Affiliation(s)
- Cecilia Evangelisti
- Medical Genetics Unit, IRCCS Azienda Ospedaliero-Universitaria di Bologna, 40138 Bologna, Italy
| | - Sherin Ramadan
- Department of Medical and Surgical Sciences, University of Bologna, 40138 Bologna, Italy
| | - Antonio Orlacchio
- Department of Medicine and Surgery, University of Perugia, 06123 Perugia, Italy
- Laboratory of Neurogenetics, European Center for Brain Research (CERC), IRCCS Santa Lucia Foundation, 00143 Rome, Italy
| | - Emanuele Panza
- Medical Genetics Unit, IRCCS Azienda Ospedaliero-Universitaria di Bologna, 40138 Bologna, Italy
- Department of Medical and Surgical Sciences, University of Bologna, 40138 Bologna, Italy
| |
Collapse
|
33
|
Gutierrez-Barbosa H, Medina-Moreno S, Perdomo-Celis F, Davis H, Chua JV, Zapata JC. Evaluation of Four Humanized NOD-Derived Mouse Models for Dengue Virus-2 Infection. Pathogens 2024; 13:639. [PMID: 39204240 PMCID: PMC11357684 DOI: 10.3390/pathogens13080639] [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: 06/26/2024] [Revised: 07/25/2024] [Accepted: 07/29/2024] [Indexed: 09/03/2024] Open
Abstract
Dengue is a significant public health problem with no specific viral treatment. One of the main challenges in studying dengue is the lack of adequate animal models recapitulating human immune responses. Most studies on humanized mice use NOD-scid IL2R gamma null (NSG) mice, which exhibit poor hematopoiesis for some cell populations. This study compares three humanized (hu) NOD-derived mouse models for dengue virus-2 (DENV-2) infection in the context of human cytokine expression. Three mouse strains (hu-NSG, hu-EXL, and hu-SGM3) received xenotransplants of human CD34+ fetal cord blood cells from a single donor, and one mouse strain received human peripheral blood mononuclear cells (hu-SGM3-PBMCs). All models exhibited infectious viruses in blood confirmed by plaque assay, but mice expressing human cytokines showed higher viremia compared to conventional NSG mice. The hu-SGM3-PBMCs model developed lethal infections, showing a significant increase in viremia and clinical signs. A detectable human cytokine response was observed in all the DENV-2-infected humanized mouse models. In conclusion, humanized NOD-derived mouse models expressing human cytokines offer a relevant platform for the study of dengue pathogenesis and antiviral therapies.
Collapse
Affiliation(s)
- Hernando Gutierrez-Barbosa
- Institute of Human Virology, University of Maryland School of Medicine, Baltimore, MD 21201, USA; (H.G.-B.); (S.M.-M.); (J.V.C.)
- Facultad de Biología, Universidad de Antioquia, Bogotá 050010, Colombia
| | - Sandra Medina-Moreno
- Institute of Human Virology, University of Maryland School of Medicine, Baltimore, MD 21201, USA; (H.G.-B.); (S.M.-M.); (J.V.C.)
| | - Federico Perdomo-Celis
- Instituto de Genética Humana, Facultad de Medicina, Pontificia Universidad Javeriana, Bogotá 110231, Colombia;
| | - Harry Davis
- Institute of Human Virology, University of Maryland School of Medicine, Baltimore, MD 21201, USA; (H.G.-B.); (S.M.-M.); (J.V.C.)
| | - Joel V. Chua
- Institute of Human Virology, University of Maryland School of Medicine, Baltimore, MD 21201, USA; (H.G.-B.); (S.M.-M.); (J.V.C.)
| | - Juan C. Zapata
- Institute of Human Virology, University of Maryland School of Medicine, Baltimore, MD 21201, USA; (H.G.-B.); (S.M.-M.); (J.V.C.)
| |
Collapse
|
34
|
Liu T, Li M, Tian Y, Dong Y, Liu N, Wang Z, Zhang H, Zheng A, Cui C. Immunogenicity and safety of a self-assembling ZIKV nanoparticle vaccine in mice. Int J Pharm 2024; 660:124320. [PMID: 38866086 DOI: 10.1016/j.ijpharm.2024.124320] [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: 02/14/2024] [Revised: 05/07/2024] [Accepted: 06/06/2024] [Indexed: 06/14/2024]
Abstract
Zika virus (ZIKV) is a mosquito-borne flavivirus that highly susceptibly causes Guillain-Barré syndrome and microcephaly in newborns. Vaccination is one of the most effective measures for preventing infectious diseases. However, there is currently no approved vaccine to prevent ZIKV infection. Here, we developed nanoparticle (NP) vaccines by covalently conjugating self-assembled 24-subunit ferritin to the envelope structural protein subunit of ZIKV to achieve antigen polyaggregation. The immunogenicityof the NP vaccine was evaluated in mice. Compared to monomer vaccines, the NP vaccine achieved effective antigen presentation, promoted the differentiation of follicular T helper cells in lymph nodes, and induced significantly greater antigen-specific humoral and cellular immune responses. Moreover, the NP vaccine enhanced high-affinity antigen-specific IgG antibody levels, increased secretion of the cytokines IL-4 and IFN-γ by splenocytes, significantly activated T/B lymphocytes, and improved the generation of memory T/B cells. In addition, no significant adverse reactions occurred when NP vaccine was combined with adjuvants. Overall, ferritin-based NP vaccines are safe and effective ZIKV vaccine candidates.
Collapse
Affiliation(s)
- Ting Liu
- Department of Pharmaceutics, School of Pharmaceutical Sciences, Capital Medical University, Beijing 100069, China; Beijing Area Major Laboratory of Peptide and Small Molecular Drugs, Beijing 100069, China; Engineering Research Center of Endogenous Prophylactic of Ministry of Education of China, Beijing 100069, China; Beijing Laboratory of Biomedical Materials, Beijing 100069, China
| | - Meng Li
- State Key Laboratory of Toxicology and Medical Countermeasures, Beijing Institute of Pharmacology and Toxicology, Beijing 100850, China
| | - Yang Tian
- Department of Pharmaceutics, School of Pharmaceutical Sciences, Capital Medical University, Beijing 100069, China; Beijing Area Major Laboratory of Peptide and Small Molecular Drugs, Beijing 100069, China; Engineering Research Center of Endogenous Prophylactic of Ministry of Education of China, Beijing 100069, China; Beijing Laboratory of Biomedical Materials, Beijing 100069, China
| | - Yuhan Dong
- State Key Laboratory of Toxicology and Medical Countermeasures, Beijing Institute of Pharmacology and Toxicology, Beijing 100850, China
| | - Nan Liu
- State Key Laboratory of Toxicology and Medical Countermeasures, Beijing Institute of Pharmacology and Toxicology, Beijing 100850, China
| | - Zengming Wang
- State Key Laboratory of Toxicology and Medical Countermeasures, Beijing Institute of Pharmacology and Toxicology, Beijing 100850, China
| | - Hui Zhang
- State Key Laboratory of Toxicology and Medical Countermeasures, Beijing Institute of Pharmacology and Toxicology, Beijing 100850, China
| | - Aiping Zheng
- State Key Laboratory of Toxicology and Medical Countermeasures, Beijing Institute of Pharmacology and Toxicology, Beijing 100850, China
| | - Chunying Cui
- Department of Pharmaceutics, School of Pharmaceutical Sciences, Capital Medical University, Beijing 100069, China; Beijing Area Major Laboratory of Peptide and Small Molecular Drugs, Beijing 100069, China; Engineering Research Center of Endogenous Prophylactic of Ministry of Education of China, Beijing 100069, China; Beijing Laboratory of Biomedical Materials, Beijing 100069, China.
| |
Collapse
|
35
|
Kan H, Chen Y. Revealing endogenous conditions for Peto's paradox via an ordinary differential equation model. J Math Biol 2024; 89:27. [PMID: 38970664 PMCID: PMC11227477 DOI: 10.1007/s00285-024-02123-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2023] [Revised: 06/21/2024] [Accepted: 06/24/2024] [Indexed: 07/08/2024]
Abstract
Cancer, a disease intimately linked to cellular mutations, is commonly believed to exhibit a positive association with the cell count and lifespan of a species. Despite this assumption, the observed uniformity in cancer rates across species, referred to as the Peto's paradox, presents a conundrum. Recognizing that tumour progression is not solely dependent on cancer cells but involves intricate interactions among various cell types, this study employed a Lotka-Volterra (LV) ordinary differential equation model to analyze the evolution of cancerous cells and the cancer incidence in an immune environment. As a result, this study uncovered the sufficient conditions underlying the absence of correlation in Peto's paradox and provide insights into the reasons for the equitable distribution of cancer incidence across diverse species by applying nondimensionalization and drawing an analogy between the characteristic time interval for the variation of cell populations in the ODE model and that of cell cycles of a species.
Collapse
Affiliation(s)
- Haichun Kan
- SCS Laboratory, Department of Human and Engineered Environmental Studies, Graduate School of Frontier Sciences, The University of Tokyo, Chiba, Japan
| | - Yu Chen
- SCS Laboratory, Department of Human and Engineered Environmental Studies, Graduate School of Frontier Sciences, The University of Tokyo, Chiba, Japan.
| |
Collapse
|
36
|
Love JR, Karthaus WR. Next-Generation Modeling of Cancer Using Organoids. Cold Spring Harb Perspect Med 2024; 14:a041380. [PMID: 37734867 PMCID: PMC11146310 DOI: 10.1101/cshperspect.a041380] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/23/2023]
Abstract
In the last decade, organoid technology has become a cornerstone in cancer research. Organoids are long-term primary cell cultures, usually of epithelial origin, grown in a three-dimensional (3D) protein matrix and a fully defined medium. Organoids can be derived from many organs and cancer types and sites, encompassing both murine and human tissues. Importantly, they can be established from various stages during tumor evolution and recapitulate with high accuracy patient genomics and phenotypes in vitro, offering a platform for personalized medicine. Additionally, organoids are remarkably amendable for experimental manipulation. Taken together, these features make organoids a powerful tool with applications in basic cancer research and personalized medicine. Here, we will discuss the origins of organoid culture, applications in cancer research, and how cancer organoids can synergize with other models of cancer to drive basic discoveries as well as to translate these toward clinical solutions.
Collapse
Affiliation(s)
- Jillian R Love
- Swiss Institute for Experimental Cancer Research (ISREC), School of Life Sciences, EPFL, 1015 Lausanne, Switzerland
| | - Wouter R Karthaus
- Swiss Institute for Experimental Cancer Research (ISREC), School of Life Sciences, EPFL, 1015 Lausanne, Switzerland
| |
Collapse
|
37
|
Obare LM, Bonami RH, Doran A, Wanjalla CN. B cells and atherosclerosis: A HIV perspective. J Cell Physiol 2024; 239:e31270. [PMID: 38651687 PMCID: PMC11209796 DOI: 10.1002/jcp.31270] [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: 12/04/2023] [Revised: 03/09/2024] [Accepted: 03/27/2024] [Indexed: 04/25/2024]
Abstract
Atherosclerosis remains a leading cause of cardiovascular disease (CVD) globally, with the complex interplay of inflammation and lipid metabolism at its core. Recent evidence suggests a role of B cells in the pathogenesis of atherosclerosis; however, this relationship remains poorly understood, particularly in the context of HIV. We review the multifaceted functions of B cells in atherosclerosis, with a specific focus on HIV. Unique to atherosclerosis is the pivotal role of natural antibodies, particularly those targeting oxidized epitopes abundant in modified lipoproteins and cellular debris. B cells can exert control over cellular immune responses within atherosclerotic arteries through antigen presentation, chemokine production, cytokine production, and cell-cell interactions, actively participating in local and systemic immune responses. We explore how HIV, characterized by chronic immune activation and dysregulation, influences B cells in the context of atherosclerosis, potentially exacerbating CVD risk in persons with HIV. By examining the proatherogenic and antiatherogenic properties of B cells, we aim to deepen our understanding of how B cells influence atherosclerotic plaque development, especially within the framework of HIV. This research provides a foundation for novel B cell-targeted interventions, with the potential to mitigate inflammation-driven cardiovascular events, offering new perspectives on CVD risk management in PLWH.
Collapse
Affiliation(s)
- Laventa M. Obare
- Division of Infectious Diseases, Department of Medicine, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Rachel H. Bonami
- Division of Rheumatology and Immunology, Department of Medicine, Vanderbilt University Medical Center, Nashville, TN, USA
- Department of Pathology, Microbiology, and Immunology, Vanderbilt University Medical Center, Nashville, TN, USA
- Vanderbilt Center for Immunobiology, Vanderbilt University Medical Center, Nashville, TN, USA
- Vanderbilt Institute for Infection, Immunology, and Inflammation, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Amanda Doran
- Department of Pathology, Microbiology, and Immunology, Vanderbilt University Medical Center, Nashville, TN, USA
- Vanderbilt Center for Immunobiology, Vanderbilt University Medical Center, Nashville, TN, USA
- Vanderbilt Institute for Infection, Immunology, and Inflammation, Vanderbilt University Medical Center, Nashville, TN, USA
- Division of Cardiology, Department of Medicine, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Celestine N. Wanjalla
- Division of Infectious Diseases, Department of Medicine, Vanderbilt University Medical Center, Nashville, TN, USA
- Vanderbilt Institute for Infection, Immunology, and Inflammation, Vanderbilt University Medical Center, Nashville, TN, USA
| |
Collapse
|
38
|
Huayamares SG, Loughrey D, Kim H, Dahlman JE, Sorscher EJ. Nucleic acid-based drugs for patients with solid tumours. Nat Rev Clin Oncol 2024; 21:407-427. [PMID: 38589512 DOI: 10.1038/s41571-024-00883-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/18/2024] [Indexed: 04/10/2024]
Abstract
The treatment of patients with advanced-stage solid tumours typically involves a multimodality approach (including surgery, chemotherapy, radiotherapy, targeted therapy and/or immunotherapy), which is often ultimately ineffective. Nucleic acid-based drugs, either as monotherapies or in combination with standard-of-care therapies, are rapidly emerging as novel treatments capable of generating responses in otherwise refractory tumours. These therapies include those using viral vectors (also referred to as gene therapies), several of which have now been approved by regulatory agencies, and nanoparticles containing mRNAs and a range of other nucleotides. In this Review, we describe the development and clinical activity of viral and non-viral nucleic acid-based treatments, including their mechanisms of action, tolerability and available efficacy data from patients with solid tumours. We also describe the effects of the tumour microenvironment on drug delivery for both systemically administered and locally administered agents. Finally, we discuss important trends resulting from ongoing clinical trials and preclinical testing, and manufacturing and/or stability considerations that are expected to underpin the next generation of nucleic acid agents for patients with solid tumours.
Collapse
Affiliation(s)
- Sebastian G Huayamares
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology, Atlanta, GA, USA
- Emory University School of Medicine, Atlanta, GA, USA
| | - David Loughrey
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology, Atlanta, GA, USA
- Emory University School of Medicine, Atlanta, GA, USA
| | - Hyejin Kim
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology, Atlanta, GA, USA
- Emory University School of Medicine, Atlanta, GA, USA
| | - James E Dahlman
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology, Atlanta, GA, USA.
- Emory University School of Medicine, Atlanta, GA, USA.
| | - Eric J Sorscher
- Emory University School of Medicine, Atlanta, GA, USA.
- Department of Pediatrics, Emory University, Atlanta, GA, USA.
- Winship Cancer Institute, Emory University, Atlanta, GA, USA.
| |
Collapse
|
39
|
G. de Castro C, G. del Hierro A, H-Vázquez J, Cuesta-Sancho S, Bernardo D. State-of-the-art cytometry in the search of novel biomarkers in digestive cancers. Front Oncol 2024; 14:1407580. [PMID: 38868532 PMCID: PMC11167087 DOI: 10.3389/fonc.2024.1407580] [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: 03/26/2024] [Accepted: 05/10/2024] [Indexed: 06/14/2024] Open
Abstract
Despite that colorectal and liver cancer are among the most prevalent tumours in the world, the identification of non-invasive biomarkers to aid on their diagnose and subsequent prognosis is a current unmet need that would diminish both their incidence and mortality rates. In this context, conventional flow cytometry has been widely used in the screening of biomarkers with clinical utility in other malignant processes like leukaemia or lymphoma. Therefore, in this review, we will focus on how advanced cytometry panels covering over 40 parameters can be applied on the study of the immune system from patients with colorectal and hepatocellular carcinoma and how that can be used on the search of novel biomarkers to aid or diagnose, prognosis, and even predict clinical response to different treatments. In addition, these multiparametric and unbiased approaches can also provide novel insights into the specific immunopathogenic mechanisms governing these malignant diseases, hence potentially unravelling novel targets to perform immunotherapy or identify novel mechanisms, rendering the development of novel treatments. As a consequence, computational cytometry approaches are an emerging methodology for the early detection and predicting therapies for gastrointestinal cancers.
Collapse
Affiliation(s)
- Carolina G. de Castro
- Mucosal Immunology Lab, Institute of Biomedicine and Molecular Genetics (IBGM), University of Valladolid and Consejo Superior de Investigaciones Científicas (CSIC), Valladolid, Spain
| | - Alejandro G. del Hierro
- Mucosal Immunology Lab, Institute of Biomedicine and Molecular Genetics (IBGM), University of Valladolid and Consejo Superior de Investigaciones Científicas (CSIC), Valladolid, Spain
| | - Juan H-Vázquez
- Mucosal Immunology Lab, Institute of Biomedicine and Molecular Genetics (IBGM), University of Valladolid and Consejo Superior de Investigaciones Científicas (CSIC), Valladolid, Spain
| | - Sara Cuesta-Sancho
- Mucosal Immunology Lab, Institute of Biomedicine and Molecular Genetics (IBGM), University of Valladolid and Consejo Superior de Investigaciones Científicas (CSIC), Valladolid, Spain
| | - David Bernardo
- Mucosal Immunology Lab, Institute of Biomedicine and Molecular Genetics (IBGM), University of Valladolid and Consejo Superior de Investigaciones Científicas (CSIC), Valladolid, Spain
- Centro de Investigaciones Biomedicas en Red de Enfermedades Infecciosas (CIBERINFEC), Madrid, Spain
| |
Collapse
|
40
|
Shin DS, Basak S, Veena MS, Comin‐Anduix B, Bhattacharya A, Dong TS, Ko A, Han P, Jacobs J, Moatamed NA, Avila L, Pellegrini M, Wang M, Srivatsan ES. Enhanced CTLA-4 blockade anti-tumor immunity with APG-157 combination in a murine head and neck cancer. Cancer Med 2024; 13:e7212. [PMID: 38686626 PMCID: PMC11058674 DOI: 10.1002/cam4.7212] [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: 12/11/2023] [Revised: 03/19/2024] [Accepted: 04/11/2024] [Indexed: 05/02/2024] Open
Abstract
BACKGROUND A phase I clinical study for patients with locally advanced H&N cancer with a new class of botanical drug APG-157 provided hints of potential synergy with immunotherapy. We sought to evaluate the efficacy of the combination of APG-157 and immune checkpoint inhibitors. METHODS CCL23, UM-SCC1 (human), and SCCVII (HPV-), MEER (HPV+) (murine) H&N cancer cell lines were utilized for in vitro and in vivo studies. We measured tumor growth by treating the mice with APG-157, anti-PD-1, and anti-CTLA-4 antibody combinations (8 groups). The tumor microenvironments were assessed by multi-color flow cytometry, immunohistochemistry, and RNA-seq analysis. Fecal microbiome was analyzed by 16S rRNA sequence. RESULTS Among the eight treatment groups, APG-157 + anti-CTLA-4 demonstrated the best tumor growth suppression (p = 0.0065 compared to the control), followed by anti-PD-1 + anti-CTLA-4 treatment group (p = 0.48 compared to the control). Immunophenotype showed over 30% of CD8+ T cells in APG-157 + anti-CTLA-4 group compared to 4%-5% of CD8+ T cells for the control group. Differential gene expression analysis revealed that APG-157 + anti-CTLA-4 group showed an enriched set of genes for inflammatory response and apoptotic signaling pathways. The fecal microbiome analysis showed a substantial difference of lactobacillus genus among groups, highest for APG-157 + anti-CTLA-4 treatment group. We were unable to perform correlative studies for MEER model as there was tumor growth suppression with all treatment conditions, except for the untreated control group. CONCLUSIONS The results indicate that APG-157 and immune checkpoint inhibitor combination treatment could potentially lead to improved tumor control.
Collapse
Affiliation(s)
- Daniel Sanghoon Shin
- Department of Medicine, Division of Hematology‐OncologyVAGLAHS/David Geffen School of Medicine at University of California, Los Angeles (UCLA)Los AngelesCaliforniaUSA
| | - Saroj Basak
- Department of Medicine, Division of Hematology‐OncologyVAGLAHS/David Geffen School of Medicine at University of California, Los Angeles (UCLA)Los AngelesCaliforniaUSA
| | - Mysore S. Veena
- Department of Medicine, Division of Hematology‐OncologyVAGLAHS/David Geffen School of Medicine at University of California, Los Angeles (UCLA)Los AngelesCaliforniaUSA
| | - Begoña Comin‐Anduix
- Department of SurgeryDavid Geffen School of Medicine at UCLALos AngelesCaliforniaUSA
- Parker Institute for Cancer ImmunotherapyLos AngelesCaliforniaUSA
- Ahmanson Translational Theranostics Division, Department of Molecular and Medical PharmacologyUCLALos Angeles, CaliforniaUSA
| | - Arjun Bhattacharya
- Department of Chemistry and Biochemistry and the Institute for Quantitative and Computational BiologyUCLALos AngelesCaliforniaUSA
- Department of Louise M. Darling Biomedical Library and The Institute for Quantitative and Computational BiologyUCLALos AngelesCaliforniaUSA
- Present address:
Department of EpidemiologyThe University of Texas MD Anderson Cancer CenterHoustonTexasUSA
| | - Tien S. Dong
- Division of Digestive Diseases, Department of MedicineDavid Geffen School of Medicine at UCLALos AngelesCaliforniaUSA
| | - Albert Ko
- Department of SurgeryDavid Geffen School of Medicine at UCLALos AngelesCaliforniaUSA
| | - Philip Han
- Department of SurgeryDavid Geffen School of Medicine at UCLALos AngelesCaliforniaUSA
| | - Jonathan Jacobs
- Division of Digestive Diseases, Department of MedicineDavid Geffen School of Medicine at UCLALos AngelesCaliforniaUSA
| | - Neda A. Moatamed
- Department of Pathology and Laboratory MedicineDavid Geffen School of Medicine at UCLALos AngelesCaliforniaUSA
| | | | - Matteo Pellegrini
- Department of Chemistry and Biochemistry and the Institute for Quantitative and Computational BiologyUCLALos AngelesCaliforniaUSA
- Department of Louise M. Darling Biomedical Library and The Institute for Quantitative and Computational BiologyUCLALos AngelesCaliforniaUSA
- Department of MolecularCellular and Developmental Biology, UCLALos AngelesCaliforniaUSA
| | - Marilene Wang
- Department of Surgery, VAGLAHS and Department of Head and Neck SurgeryDavid Geffen School of Medicine at UCLALos AngelesCaliforniaUSA
| | - Eri S. Srivatsan
- Department of SurgeryVAGLAHS/David Geffen School of Medicine at UCLALos AngelesCaliforniaUSA
| |
Collapse
|
41
|
Gardet M, Haigh O, Meurisse F, Coindre S, Dimant N, Desjardins D, Bourgeois C, Goujard C, Vaslin B, Relouzat F, Le Grand R, Lambotte O, Favier B. Identification of macaque dendritic cell precursors in blood and tissue reveals their dysregulation in early SIV infection. Cell Rep 2024; 43:113994. [PMID: 38530856 DOI: 10.1016/j.celrep.2024.113994] [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: 04/14/2023] [Revised: 01/27/2024] [Accepted: 03/08/2024] [Indexed: 03/28/2024] Open
Abstract
Distinct dendritic cell (DC) subsets play important roles in shaping immune responses. Circulating DC precursors (pre-DCs) are more susceptible to HIV infection in vitro, which may explain the inefficiency of immune responses against HIV. However, the interplay between HIV and pre-DC is not defined in vivo. We identify human pre-DC equivalents in the cynomolgus macaque and then analyze their dynamics during simian immunodeficiency virus (SIV) infection to illustrate a sharp decrease of blood pre-DCs in early SIV infection and accumulation in lymph nodes (LNs), where they neglect to upregulate CD83/CD86 or MHC-II. Additionally, SIV infection attenuates the capacity of stimulated LN pre-DCs to produce IL-12p40. Analysis of HIV cohorts provides correlation between costimulatory molecule expression on pre-DCs and T cell activation in spontaneous HIV controllers. These findings pinpoint certain dynamics and functional changes of pre-DCs during SIV infection, providing a deeper understanding of immune dysregulation mechanisms elicited in people living with HIV.
Collapse
Affiliation(s)
- Margaux Gardet
- Université Paris-Saclay, Inserm, CEA, Center for Immunology of Viral, Auto-immune, Hematological and Bacterial Diseases (IMVA-HB/IDMIT), 92265 Fontenay-aux-Roses, France
| | - Oscar Haigh
- Université Paris-Saclay, Inserm, CEA, Center for Immunology of Viral, Auto-immune, Hematological and Bacterial Diseases (IMVA-HB/IDMIT), 92265 Fontenay-aux-Roses, France
| | - Florian Meurisse
- Université Paris-Saclay, Inserm, CEA, Center for Immunology of Viral, Auto-immune, Hematological and Bacterial Diseases (IMVA-HB/IDMIT), 92265 Fontenay-aux-Roses, France
| | - Sixtine Coindre
- Université Paris-Saclay, Inserm, CEA, Center for Immunology of Viral, Auto-immune, Hematological and Bacterial Diseases (IMVA-HB/IDMIT), 92265 Fontenay-aux-Roses, France
| | - Nastasia Dimant
- Université Paris-Saclay, Inserm, CEA, Center for Immunology of Viral, Auto-immune, Hematological and Bacterial Diseases (IMVA-HB/IDMIT), 92265 Fontenay-aux-Roses, France
| | - Delphine Desjardins
- Université Paris-Saclay, Inserm, CEA, Center for Immunology of Viral, Auto-immune, Hematological and Bacterial Diseases (IMVA-HB/IDMIT), 92265 Fontenay-aux-Roses, France
| | - Christine Bourgeois
- Université Paris-Saclay, Inserm, CEA, Center for Immunology of Viral, Auto-immune, Hematological and Bacterial Diseases (IMVA-HB/IDMIT), 92265 Fontenay-aux-Roses, France
| | - Cecile Goujard
- Paris-Saclay University Hospital Group, Assistance Publique Hôpitaux de Paris, Department of Internal Medicine and Clinical Immunology, Bicêtre Hospital, le Kremlin-Bicêtre, France; Centre de Recherche en Épidémiologie et Santé des Populations (CESP), INSERM U1018, University Paris Saclay, Paris, France
| | - Bruno Vaslin
- Université Paris-Saclay, Inserm, CEA, Center for Immunology of Viral, Auto-immune, Hematological and Bacterial Diseases (IMVA-HB/IDMIT), 92265 Fontenay-aux-Roses, France
| | - Francis Relouzat
- Université Paris-Saclay, Inserm, CEA, Center for Immunology of Viral, Auto-immune, Hematological and Bacterial Diseases (IMVA-HB/IDMIT), 92265 Fontenay-aux-Roses, France
| | - Roger Le Grand
- Université Paris-Saclay, Inserm, CEA, Center for Immunology of Viral, Auto-immune, Hematological and Bacterial Diseases (IMVA-HB/IDMIT), 92265 Fontenay-aux-Roses, France
| | - Olivier Lambotte
- Université Paris-Saclay, Inserm, CEA, Center for Immunology of Viral, Auto-immune, Hematological and Bacterial Diseases (IMVA-HB/IDMIT), 92265 Fontenay-aux-Roses, France; Paris-Saclay University Hospital Group, Assistance Publique Hôpitaux de Paris, Department of Internal Medicine and Clinical Immunology, Bicêtre Hospital, le Kremlin-Bicêtre, France
| | - Benoit Favier
- Université Paris-Saclay, Inserm, CEA, Center for Immunology of Viral, Auto-immune, Hematological and Bacterial Diseases (IMVA-HB/IDMIT), 92265 Fontenay-aux-Roses, France.
| |
Collapse
|
42
|
Koca MB, Sevilgen FE. Integration of single-cell proteomic datasets through distinctive proteins in cell clusters. Proteomics 2024; 24:e2300282. [PMID: 38135888 DOI: 10.1002/pmic.202300282] [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: 07/19/2023] [Revised: 11/01/2023] [Accepted: 12/04/2023] [Indexed: 12/24/2023]
Abstract
The use of mass spectrometry and antibody-based sequencing technologies at the single-cell level has led to an increase in single-cell proteomic datasets. Integrating these datasets is crucial to eliminate the batch effect that often arises due to their limited sequencing molecules. Although methods for horizontally integrating high-dimensional single-cell transcriptomic datasets can also be applied to single-cell proteomic datasets, a specialized approach explicitly tailored for low-dimensional proteomic datasets may enhance the integration process. Here, we introduce SCPRO-HI, an algorithm for the horizontal integration of antibody-based single-cell proteomic datasets. It utilizes a hierarchical cell anchoring technique to match cells based on the similarity of distinctive proteins for constituting cell clusters. A novel variational auto-encoder model is employed for correcting batch effects on the protein abundances, eliminating the need for mapping them into a new domain. Moreover, we propose a technique for extending the algorithm to high-dimensional datasets. The performance of the SCPRO-HI algorithm is evaluated using simulated and real-world single-cell proteomic datasets. The findings demonstrate our algorithm outperforms state-of-the-art methods, achieving a 75% higher silhouette score while preserving HVPs 13% better. Furthermore, the algorithm shows competitive performance in transcriptomic datasets, suggesting potential for integrating high-dimensional mass-spectrometry-based proteomic datasets.
Collapse
Affiliation(s)
- Mehmet Burak Koca
- Computer Engineering Department, Gebze Technical University, Kocaeli, Türkiye
| | - Fatih Erdoğan Sevilgen
- Institute for Data Science and Artificial Intelligence, Boğaziçi University, İstanbul, Türkiye
| |
Collapse
|
43
|
Amartumur S, Nguyen H, Huynh T, Kim TS, Woo RS, Oh E, Kim KK, Lee LP, Heo C. Neuropathogenesis-on-chips for neurodegenerative diseases. Nat Commun 2024; 15:2219. [PMID: 38472255 PMCID: PMC10933492 DOI: 10.1038/s41467-024-46554-8] [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: 10/04/2023] [Accepted: 02/28/2024] [Indexed: 03/14/2024] Open
Abstract
Developing diagnostics and treatments for neurodegenerative diseases (NDs) is challenging due to multifactorial pathogenesis that progresses gradually. Advanced in vitro systems that recapitulate patient-like pathophysiology are emerging as alternatives to conventional animal-based models. In this review, we explore the interconnected pathogenic features of different types of ND, discuss the general strategy to modelling NDs using a microfluidic chip, and introduce the organoid-on-a-chip as the next advanced relevant model. Lastly, we overview how these models are being applied in academic and industrial drug development. The integration of microfluidic chips, stem cells, and biotechnological devices promises to provide valuable insights for biomedical research and developing diagnostic and therapeutic solutions for NDs.
Collapse
Affiliation(s)
- Sarnai Amartumur
- Department of Biophysics, Institute of Quantum Biophysics, Sungkyunkwan University, Suwon, 16419, Korea
| | - Huong Nguyen
- Department of Biophysics, Institute of Quantum Biophysics, Sungkyunkwan University, Suwon, 16419, Korea
| | - Thuy Huynh
- Department of Biophysics, Institute of Quantum Biophysics, Sungkyunkwan University, Suwon, 16419, Korea
| | - Testaverde S Kim
- Center for Integrated Nanostructure Physics (CINAP), Institute for Basic Science (IBS), Suwon, 16419, Korea
| | - Ran-Sook Woo
- Department of Anatomy and Neuroscience, College of Medicine, Eulji University, Daejeon, 34824, Korea
| | - Eungseok Oh
- Department of Neurology, Chungnam National University Hospital, Daejeon, 35015, Korea
| | - Kyeong Kyu Kim
- Department of Precision Medicine, Graduate School of Basic Medical Science (GSBMS), Institute for Anti-microbial Resistance Research and Therapeutics, Sungkyunkwan University School of Medicine, Suwon, 16419, Korea
| | - Luke P Lee
- Department of Biophysics, Institute of Quantum Biophysics, Sungkyunkwan University, Suwon, 16419, Korea.
- Harvard Medical School, Division of Engineering in Medicine and Renal Division, Department of Medicine, Brigham and Women's Hospital, Boston, MA, 02115, USA.
- Department of Bioengineering, Department of Electrical Engineering and Computer Science, University of California, Berkeley, CA, 94720, USA.
| | - Chaejeong Heo
- Department of Biophysics, Institute of Quantum Biophysics, Sungkyunkwan University, Suwon, 16419, Korea.
- Center for Integrated Nanostructure Physics (CINAP), Institute for Basic Science (IBS), Suwon, 16419, Korea.
| |
Collapse
|
44
|
Hu X, White K, Olroyd AG, DeJesus R, Dominguez AA, Dowdle WE, Friera AM, Young C, Wells F, Chu EY, Ito CE, Krishnapura H, Jain S, Ankala R, McGill TJ, Lin A, Egenberger K, Gagnon A, Michael Rukstalis J, Hogrebe NJ, Gattis C, Basco R, Millman JR, Kievit P, Davis MM, Lanier LL, Connolly AJ, Deuse T, Schrepfer S. Hypoimmune induced pluripotent stem cells survive long term in fully immunocompetent, allogeneic rhesus macaques. Nat Biotechnol 2024; 42:413-423. [PMID: 37156915 PMCID: PMC10940156 DOI: 10.1038/s41587-023-01784-x] [Citation(s) in RCA: 66] [Impact Index Per Article: 66.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2022] [Accepted: 04/06/2023] [Indexed: 05/10/2023]
Abstract
Genetic engineering of allogeneic cell therapeutics that fully prevents rejection by a recipient's immune system would abolish the requirement for immunosuppressive drugs or encapsulation and support large-scale manufacturing of off-the-shelf cell products. Previously, we generated mouse and human hypoimmune pluripotent (HIP) stem cells by depleting HLA class I and II molecules and overexpressing CD47 (B2M-/-CIITA-/-CD47+). To determine whether this strategy is successful in non-human primates, we engineered rhesus macaque HIP cells and transplanted them intramuscularly into four allogeneic rhesus macaques. The HIP cells survived unrestricted for 16 weeks in fully immunocompetent allogeneic recipients and differentiated into several lineages, whereas allogeneic wild-type cells were vigorously rejected. We also differentiated human HIP cells into endocrinologically active pancreatic islet cells and showed that they survived in immunocompetent, allogeneic diabetic humanized mice for 4 weeks and ameliorated diabetes. HIP-edited primary rhesus macaque islets survived for 40 weeks in an allogeneic rhesus macaque recipient without immunosuppression, whereas unedited islets were quickly rejected.
Collapse
Affiliation(s)
- Xiaomeng Hu
- Sana Biotechnology, Inc., South San Francisco, CA, USA
| | - Kathy White
- Sana Biotechnology, Inc., South San Francisco, CA, USA
| | - Ari G Olroyd
- Sana Biotechnology, Inc., South San Francisco, CA, USA
| | | | | | | | | | - Chi Young
- Sana Biotechnology, Inc., South San Francisco, CA, USA
| | - Frank Wells
- Sana Biotechnology, Inc., South San Francisco, CA, USA
| | - Elaine Y Chu
- Sana Biotechnology, Inc., South San Francisco, CA, USA
| | | | | | - Surbhi Jain
- Sana Biotechnology, Inc., South San Francisco, CA, USA
| | - Ramya Ankala
- Sana Biotechnology, Inc., South San Francisco, CA, USA
| | | | - August Lin
- Sana Biotechnology, Inc., South San Francisco, CA, USA
| | | | | | | | - Nathaniel J Hogrebe
- Division of Endocrinology, Metabolism and Lipid Research, Washington University School of Medicine, St. Louis, MO, USA
| | - Corie Gattis
- Sana Biotechnology, Inc., South San Francisco, CA, USA
| | - Ron Basco
- Sana Biotechnology, Inc., South San Francisco, CA, USA
| | | | - Paul Kievit
- Division of Cardiometabolic Health, Oregon National Primate Research Center, Beaverton, OR, USA
| | - Mark M Davis
- Howard Hughes Medical Institute, Institute for Immunity, Transplantation and Infection, and Department of Microbiology and Immunology, Stanford University School of Medicine, Stanford, CA, USA
| | - Lewis L Lanier
- Department of Microbiology and Immunology and the Parker Institute for Cancer Immunotherapy, University of California, San Francisco, San Francisco, CA, USA
| | - Andrew J Connolly
- Department of Pathology, University of California, San Francisco, San Francisco, CA, USA
| | - Tobias Deuse
- Transplant and Stem Cell Immunobiology (TSI) Lab, Department of Surgery, Division of Cardiothoracic Surgery, University of California, San Francisco, San Francisco, CA, USA
| | | |
Collapse
|
45
|
Mansouri M, Lam J, Sung KE. Progress in developing microphysiological systems for biological product assessment. LAB ON A CHIP 2024; 24:1293-1306. [PMID: 38230512 DOI: 10.1039/d3lc00876b] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/18/2024]
Abstract
Microphysiological systems (MPS), also known as miniaturized physiological environments, have been engineered to create and study functional tissue units capable of replicating organ-level responses in specific contexts. The MPS has the potential to provide insights about the safety, characterization, and effectiveness of medical products that are different and complementary to insights gained from traditional testing systems, which can help facilitate the transition of potential medical products from preclinical phases to clinical trials, and eventually to market. While many MPS are versatile and can be used in various applications, most of the current applications have primarily focused on drug discovery and testing. Yet, there is a limited amount of research available that demonstrates the use of MPS in assessing biological products such as cellular and gene therapies. This review paper aims to address this gap by discussing recent technical advancements in MPS and their potential for assessing biological products. We further discuss the challenges and considerations involved in successful translation of MPS into mainstream product testing.
Collapse
Affiliation(s)
- Mona Mansouri
- Cellular and Tissue Therapies Branch, Office of Cellular Therapy and Human Tissue, Center for Biologics Evaluation and Research, U.S. Food and Drug Administration, Silver Spring, MD, USA.
| | - Johnny Lam
- Cellular and Tissue Therapies Branch, Office of Cellular Therapy and Human Tissue, Center for Biologics Evaluation and Research, U.S. Food and Drug Administration, Silver Spring, MD, USA.
| | - Kyung E Sung
- Cellular and Tissue Therapies Branch, Office of Cellular Therapy and Human Tissue, Center for Biologics Evaluation and Research, U.S. Food and Drug Administration, Silver Spring, MD, USA.
| |
Collapse
|
46
|
Chi CW, Lao YH, Ahmed AHR, He S, Merghoub T, Leong KW, Wang S. Enabling continuous immune cell recirculation on a microfluidic array to study immunotherapeutic interactions in a recapitulated tumour microenvironment. LAB ON A CHIP 2024; 24:396-407. [PMID: 38180130 DOI: 10.1039/d3lc00662j] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/06/2024]
Abstract
The effects of immunotherapeutics on interactions between immune and cancer cells are modulated by multiple components in the tumour microenvironment (TME), including endothelium and tumour stroma, which provide both a physical barrier and immunosuppressive stimuli. Herein, we report a recirculating chip to enable continuous immune cell recirculation through a microfluidic cell array to include these crucial players. This system consists of a three-layered cell array (μFCA) spatially emulating the TME, with tailored fluidic circuits establishing T cell recirculation. This platform enables the study of dynamics among the TME, immune cells in a circulatory system and cancer cell responses thereof. Through this system, we found that tumour endothelium hindered T cell infiltration into the reconstructed breast cancer tumour compartment. This negative effect was alleviated when treated with anti-human PD-L1 (programmed cell death ligand 1) antibody. Another key stromal component - cancer associated fibroblasts - attenuated T cell infiltration, compared against normal fibroblasts, and led to reduced apoptotic activity in cancer cells. These results confirm the capability of our tumour-on-a-chip system in identifying some key axes to target in overcoming barriers to immunotherapy by recapitulating immune cell interactions with the reconstructed TME. Our results also attest to the feasibility of scaling up this system for high-throughput cancer immunotherapeutic screening.
Collapse
Affiliation(s)
- Chun-Wei Chi
- Department of Biomedical Engineering, CUNY - City College of New York, New York, NY, 10031, USA.
- Department of Pharmaceutical Sciences, University at Buffalo, The State University of New York, Buffalo, NY 14214, USA
| | - Yeh-Hsing Lao
- Department of Pharmaceutical Sciences, University at Buffalo, The State University of New York, Buffalo, NY 14214, USA
- Department of Biomedical Engineering, Columbia University, New York, NY 10027, USA
| | - A H Rezwanuddin Ahmed
- Department of Biomedical Engineering, CUNY - City College of New York, New York, NY, 10031, USA.
| | - Siyu He
- Department of Biomedical Engineering, Columbia University, New York, NY 10027, USA
| | - Taha Merghoub
- Weill Cornell Medical College, New York, NY 10065, USA
| | - Kam W Leong
- Department of Biomedical Engineering, Columbia University, New York, NY 10027, USA
- Department of Systems Biology, Columbia University Medical Center, New York, NY 10032, USA
| | - Sihong Wang
- Department of Biomedical Engineering, CUNY - City College of New York, New York, NY, 10031, USA.
| |
Collapse
|
47
|
Hasan T, Pasala AR, Hassan D, Hanotaux J, Allan DS, Maganti HB. Homing and Engraftment of Hematopoietic Stem Cells Following Transplantation: A Pre-Clinical Perspective. Curr Oncol 2024; 31:603-616. [PMID: 38392038 PMCID: PMC10888387 DOI: 10.3390/curroncol31020044] [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: 12/22/2023] [Revised: 01/17/2024] [Accepted: 01/19/2024] [Indexed: 02/24/2024] Open
Abstract
Hematopoietic stem-cell (HSC) transplantation (HSCT) is used to treat various hematologic disorders. Use of genetically modified mouse models of hematopoietic cell transplantation has been critical in our fundamental understanding of HSC biology and in developing approaches for human patients. Pre-clinical studies in animal models provide insight into the journey of transplanted HSCs from infusion to engraftment in bone-marrow (BM) niches. Various signaling molecules and growth factors secreted by HSCs and the niche microenvironment play critical roles in homing and engraftment of the transplanted cells. The sustained equilibrium of these chemical and biologic factors ensures that engrafted HSCs generate healthy and durable hematopoiesis. Transplanted healthy HSCs compete with residual host cells to repopulate stem-cell niches in the marrow. Stem-cell niches, in particular, can be altered by the effects of previous treatments, aging, and the paracrine effects of leukemic cells, which create inhospitable bone-marrow niches that are unfavorable for healthy hematopoiesis. More work to understand how stem-cell niches can be restored to favor normal hematopoiesis may be key to reducing leukemic relapses following transplant.
Collapse
Affiliation(s)
- Tanvir Hasan
- Canadian Blood Services, Stem Cells and Centre for Innovation, Ottawa, ON K1G 4J5, Canada; (T.H.); (A.R.P.); (D.H.); (J.H.)
| | - Ajay Ratan Pasala
- Canadian Blood Services, Stem Cells and Centre for Innovation, Ottawa, ON K1G 4J5, Canada; (T.H.); (A.R.P.); (D.H.); (J.H.)
- Department of Biochemistry, Microbiology and Immunology, Faculty of Medicine, University of Ottawa, Ottawa, ON K1H 8L6, Canada
| | - Dhuha Hassan
- Canadian Blood Services, Stem Cells and Centre for Innovation, Ottawa, ON K1G 4J5, Canada; (T.H.); (A.R.P.); (D.H.); (J.H.)
| | - Justine Hanotaux
- Canadian Blood Services, Stem Cells and Centre for Innovation, Ottawa, ON K1G 4J5, Canada; (T.H.); (A.R.P.); (D.H.); (J.H.)
| | - David S. Allan
- Canadian Blood Services, Stem Cells and Centre for Innovation, Ottawa, ON K1G 4J5, Canada; (T.H.); (A.R.P.); (D.H.); (J.H.)
- Department of Biochemistry, Microbiology and Immunology, Faculty of Medicine, University of Ottawa, Ottawa, ON K1H 8L6, Canada
- Clinical Epidemiology & Regenerative Medicine, Ottawa Hospital Research Institute, Ottawa, ON K1Y 4E9, Canada
| | - Harinad B. Maganti
- Canadian Blood Services, Stem Cells and Centre for Innovation, Ottawa, ON K1G 4J5, Canada; (T.H.); (A.R.P.); (D.H.); (J.H.)
- Department of Biochemistry, Microbiology and Immunology, Faculty of Medicine, University of Ottawa, Ottawa, ON K1H 8L6, Canada
| |
Collapse
|
48
|
Woellner-Santos D, Tahira AC, Malvezzi JVM, Mesel V, Morales-Vicente DA, Trentini MM, Marques-Neto LM, Matos IA, Kanno AI, Pereira ASA, Teixeira AAR, Giordano RJ, Leite LCC, Pereira CAB, DeMarco R, Amaral MS, Verjovski-Almeida S. Schistosoma mansoni vaccine candidates identified by unbiased phage display screening in self-cured rhesus macaques. NPJ Vaccines 2024; 9:5. [PMID: 38177171 PMCID: PMC10767053 DOI: 10.1038/s41541-023-00803-x] [Citation(s) in RCA: 11] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2023] [Accepted: 11/30/2023] [Indexed: 01/06/2024] Open
Abstract
Schistosomiasis, a challenging neglected tropical disease, affects millions of people worldwide. Developing a prophylactic vaccine against Schistosoma mansoni has been hindered by the parasite's biological complexity. In this study, we utilized the innovative phage-display immunoprecipitation followed by a sequencing approach (PhIP-Seq) to screen the immune response of 10 infected rhesus macaques during self-cure and challenge-resistant phases, identifying vaccine candidates. Our high-throughput S. mansoni synthetic DNA phage-display library encoded 99.6% of 119,747 58-mer peptides, providing comprehensive coverage of the parasite's proteome. Library screening with rhesus macaques' antibodies, from the early phase of establishment of parasite infection, identified significantly enriched epitopes of parasite extracellular proteins known to be expressed in the digestive tract, shifting towards intracellular proteins during the late phase of parasite clearance. Immunization of mice with a selected pool of PhIP-Seq-enriched phage-displayed peptides from MEG proteins, cathepsins B, and asparaginyl endopeptidase significantly reduced worm burden in a vaccination assay. These findings enhance our understanding of parasite-host immune responses and provide promising prospects for developing an effective schistosomiasis vaccine.
Collapse
Affiliation(s)
- Daisy Woellner-Santos
- Laboratório de Ciclo Celular, Instituto Butantan, São Paulo, SP, Brazil
- Instituto de Química, Universidade de São Paulo, São Paulo, SP, Brazil
| | - Ana C Tahira
- Laboratório de Ciclo Celular, Instituto Butantan, São Paulo, SP, Brazil
| | - João V M Malvezzi
- Laboratório de Ciclo Celular, Instituto Butantan, São Paulo, SP, Brazil
- Instituto de Matemática e Estatística, Universidade de São Paulo, São Paulo, SP, Brazil
| | - Vinicius Mesel
- Laboratório de Ciclo Celular, Instituto Butantan, São Paulo, SP, Brazil
| | - David A Morales-Vicente
- Laboratório de Ciclo Celular, Instituto Butantan, São Paulo, SP, Brazil
- Instituto de Química, Universidade de São Paulo, São Paulo, SP, Brazil
| | - Monalisa M Trentini
- Laboratório de Desenvolvimento de Vacinas, Instituto Butantan, São Paulo, SP, Brazil
| | - Lázaro M Marques-Neto
- Laboratório de Desenvolvimento de Vacinas, Instituto Butantan, São Paulo, SP, Brazil
| | - Isaac A Matos
- Instituto de Química, Universidade de São Paulo, São Paulo, SP, Brazil
| | - Alex I Kanno
- Laboratório de Desenvolvimento de Vacinas, Instituto Butantan, São Paulo, SP, Brazil
| | - Adriana S A Pereira
- Laboratório de Ciclo Celular, Instituto Butantan, São Paulo, SP, Brazil
- Instituto de Química, Universidade de São Paulo, São Paulo, SP, Brazil
| | - André A R Teixeira
- Instituto de Química, Universidade de São Paulo, São Paulo, SP, Brazil
- Institute for Protein Innovation, Boston, MA, USA
| | | | - Luciana C C Leite
- Laboratório de Desenvolvimento de Vacinas, Instituto Butantan, São Paulo, SP, Brazil
| | - Carlos A B Pereira
- Instituto de Matemática e Estatística, Universidade de São Paulo, São Paulo, SP, Brazil
| | - Ricardo DeMarco
- Instituto de Física de São Carlos, Universidade de São Paulo, São Carlos, SP, Brazil
| | - Murilo S Amaral
- Laboratório de Ciclo Celular, Instituto Butantan, São Paulo, SP, Brazil
| | - Sergio Verjovski-Almeida
- Laboratório de Ciclo Celular, Instituto Butantan, São Paulo, SP, Brazil.
- Instituto de Química, Universidade de São Paulo, São Paulo, SP, Brazil.
| |
Collapse
|
49
|
Emborg ME, Gambardella JC, Zhang A, Federoff HJ. Autologous vs heterologous cell replacement strategies for Parkinson disease and other neurologic diseases. HANDBOOK OF CLINICAL NEUROLOGY 2024; 205:41-56. [PMID: 39341662 DOI: 10.1016/b978-0-323-90120-8.00010-1] [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] [Indexed: 10/01/2024]
Abstract
Successful cell replacement strategies for brain repair depend on graft integration into the neural network, which is affected by the immune response to the grafted cells. Using Parkinson disease as an example, in this chapter, we consider the immune system interaction and its role in autologous vs heterologous graft survival and integration, as well as past and emerging strategies to overcome the immunologic response. We also reflect on the role of nonhuman primate research to assess novel approaches and consider the role of different stakeholders on advancing the most promising new approaches into the clinic.
Collapse
Affiliation(s)
- Marina E Emborg
- Preclinical Parkinson's Research Program, Wisconsin National Primate Research Center, University of Wisconsin-Madison, Madison, WI, United States; Cellular and Molecular Pathology Graduate Program, University of Wisconsin-Madison, Madison, WI, United States; Department of Medical Physics, University of Wisconsin-Madison, Madison, WI, United States.
| | - Julia C Gambardella
- Preclinical Parkinson's Research Program, Wisconsin National Primate Research Center, University of Wisconsin-Madison, Madison, WI, United States; Cellular and Molecular Pathology Graduate Program, University of Wisconsin-Madison, Madison, WI, United States
| | - Ai Zhang
- Aspen Neuroscience, San Diego, CA, United States
| | - Howard J Federoff
- Kenai Therapeutics, San Diego, CA, United States; Georgetown University Medical Center, Georgetown, Washington, DC, United States
| |
Collapse
|
50
|
Gehl V, O'Rourke CJ, Andersen JB. Immunogenomics of cholangiocarcinoma. Hepatology 2023:01515467-990000000-00649. [PMID: 37972940 DOI: 10.1097/hep.0000000000000688] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/01/2023] [Accepted: 10/16/2023] [Indexed: 11/19/2023]
Abstract
The development of cholangiocarcinoma spans years, if not decades, during which the immune system becomes corrupted and permissive to primary tumor development and metastasis. This involves subversion of local immunity at tumor sites, as well as systemic immunity and the wider host response. While immune dysfunction is a hallmark of all cholangiocarcinoma, the specific steps of the cancer-immunity cycle that are perturbed differ between patients. Heterogeneous immune functionality impacts the evolutionary development, pathobiological behavior, and therapeutic response of these tumors. Integrative genomic analyses of thousands of primary tumors have supported a biological rationale for immune-based stratification of patients, encompassing immune cell composition and functionality. However, discerning immune alterations responsible for promoting tumor initiation, maintenance, and progression from those present as bystander events remains challenging. Functionally uncoupling the tumor-promoting or tumor-suppressing roles of immune profiles will be critical for identifying new immunomodulatory treatment strategies and associated biomarkers for patient stratification. This review will discuss the immunogenomics of cholangiocarcinoma, including the impact of genomic alterations on immune functionality, subversion of the cancer-immunity cycle, as well as clinical implications for existing and novel treatment strategies.
Collapse
Affiliation(s)
- Virag Gehl
- Department of Health and Medical Sciences, Biotech Research and Innovation Centre (BRIC), University of Copenhagen, Copenhagen, Denmark
| | | | | |
Collapse
|