1
|
Sillau SH, Coughlan C, Ahmed MM, Nair K, Araya P, Galbraith MD, Bettcher BM, Espinosa JM, Chial HJ, Epperson N, Boyd TD, Potter H. Neuron loss in the brain starts in childhood, increases exponentially with age and is halted by GM-CSF treatment in Alzheimer's disease. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2024:2024.07.14.24310223. [PMID: 39072024 PMCID: PMC11275665 DOI: 10.1101/2024.07.14.24310223] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/30/2024]
Abstract
Aging increases the risk of neurodegeneration, cognitive decline, and Alzheimer's disease (AD). Currently no means exist to measure neuronal cell death during life or to prevent it. Here we show that cross-sectional measures of human plasma proteins released from dying/damaged neurons (ubiquitin C-terminal hydrolase-L1/UCH-L1 and neurofilament light/NfL) become exponentially higher from age 2-85; UCH-L1 rises faster in females. Glial fibrillary acidic protein (GFAP) concentrations, indicating astrogliosis/inflammation, increase exponentially after age 40. Treatment with human granulocyte-macrophage colony-stimulating factor (GM-CSF/sargramostim) halted neuronal cell death, as evidenced by reduced plasma UCH-L1 concentrations, in AD participants to levels equivalent to those of five-year-old healthy controls. The ability of GM-CSF treatment to reduce neuronal apoptosis was confirmed in a rat model of AD. These findings suggest that the exponential increase in neurodegeneration with age, accelerated by neuroinflammation, may underlie the contribution of aging to cognitive decline and AD and can be halted by GM-CSF/sargramostim treatment.
Collapse
|
2
|
Tobeh NS, Bruce KD. Emerging Alzheimer's disease therapeutics: promising insights from lipid metabolism and microglia-focused interventions. Front Aging Neurosci 2023; 15:1259012. [PMID: 38020773 PMCID: PMC10630922 DOI: 10.3389/fnagi.2023.1259012] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2023] [Accepted: 10/03/2023] [Indexed: 12/01/2023] Open
Abstract
More than 55 million people suffer from dementia, with this number projected to double every 20 years. In the United States, 1 in 3 aged individuals dies from Alzheimer's disease (AD) or another type of dementia and AD kills more individuals than breast cancer and prostate cancer combined. AD is a complex and multifactorial disease involving amyloid plaque and neurofibrillary tangle formation, glial cell dysfunction, and lipid droplet accumulation (among other pathologies), ultimately leading to neurodegeneration and neuronal death. Unfortunately, the current FDA-approved therapeutics do not reverse nor halt AD. While recently approved amyloid-targeting antibodies can slow AD progression to improve outcomes for some patients, they are associated with adverse side effects, may have a narrow therapeutic window, and are expensive. In this review, we evaluate current and emerging AD therapeutics in preclinical and clinical development and provide insight into emerging strategies that target brain lipid metabolism and microglial function - an approach that may synergistically target multiple mechanisms that drive AD neuropathogenesis. Overall, we evaluate whether these disease-modifying emerging therapeutics hold promise as interventions that may be able to reverse or halt AD progression.
Collapse
Affiliation(s)
- Nour S Tobeh
- Division of Endocrinology, Metabolism and Diabetes, Department of Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO, United States
- Skaggs School of Pharmacy and Pharmaceutical Sciences, University of Colorado Anschutz Medical Campus, Aurora, CO, United States
| | - Kimberley D Bruce
- Division of Endocrinology, Metabolism and Diabetes, Department of Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO, United States
| |
Collapse
|
3
|
Melchiorri D, Merlo S, Micallef B, Borg JJ, Dráfi F. Alzheimer's disease and neuroinflammation: will new drugs in clinical trials pave the way to a multi-target therapy? Front Pharmacol 2023; 14:1196413. [PMID: 37332353 PMCID: PMC10272781 DOI: 10.3389/fphar.2023.1196413] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2023] [Accepted: 05/02/2023] [Indexed: 06/20/2023] Open
Abstract
Despite extensive research, no disease-modifying therapeutic option, able to prevent, cure or halt the progression of Alzheimer's disease [AD], is currently available. AD, a devastating neurodegenerative pathology leading to dementia and death, is characterized by two pathological hallmarks, the extracellular deposits of amyloid beta (Aβ) and the intraneuronal deposits of neurofibrillary tangles (NFTs) consisting of altered hyperphosphorylated tau protein. Both have been widely studied and pharmacologically targeted for many years, without significant therapeutic results. In 2022, positive data on two monoclonal antibodies targeting Aβ, donanemab and lecanemab, followed by the 2023 FDA accelerated approval of lecanemab and the publication of the final results of the phase III Clarity AD study, have strengthened the hypothesis of a causal role of Aβ in the pathogenesis of AD. However, the magnitude of the clinical effect elicited by the two drugs is limited, suggesting that additional pathological mechanisms may contribute to the disease. Cumulative studies have shown inflammation as one of the main contributors to the pathogenesis of AD, leading to the recognition of a specific role of neuroinflammation synergic with the Aβ and NFTs cascades. The present review provides an overview of the investigational drugs targeting neuroinflammation that are currently in clinical trials. Moreover, their mechanisms of action, their positioning in the pathological cascade of events that occur in the brain throughout AD disease and their potential benefit/limitation in the therapeutic strategy in AD are discussed and highlighted as well. In addition, the latest patent requests for inflammation-targeting therapeutics to be developed in AD will also be discussed.
Collapse
Affiliation(s)
- Daniela Melchiorri
- Department of Physiology and Pharmacology, Sapienza University, Rome, Italy
| | - Sara Merlo
- Department of Biomedical and Biotechnological Sciences, Section of Pharmacology, University of Catania, Catania, Italy
| | | | - John-Joseph Borg
- Malta Medicines Authority, San Ġwann, Malta
- School of Pharmacy, Department of Biology, University of Tor Vergata, Rome, Italy
| | - František Dráfi
- Institute of Experimental Pharmacology and Toxicology, Centre of Experimental Medicine SAS Bratislava, Bratislava, Slovakia
- State Institute for Drug Control, Bratislava, Slovakia
| |
Collapse
|
4
|
Stonedahl S, Leser JS, Clarke P, Potter H, Boyd TD, Tyler KL. Treatment with Granulocyte-Macrophage Colony-Stimulating Factor Reduces Viral Titers in the Brains of West Nile Virus-Infected Mice and Improves Survival. J Virol 2023; 97:e0180522. [PMID: 36802227 PMCID: PMC10062152 DOI: 10.1128/jvi.01805-22] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2022] [Accepted: 01/29/2023] [Indexed: 02/23/2023] Open
Abstract
West Nile virus (WNV) is the leading cause of epidemic arboviral encephalitis in the United States. As there are currently no proven antiviral therapies or licensed human vaccines, understanding the neuropathogenesis of WNV is critical for rational therapeutic design. In WNV-infected mice, the depletion of microglia leads to enhanced viral replication, increased central nervous system (CNS) tissue injury, and increased mortality, suggesting that microglia play a critical role in protection against WNV neuroinvasive disease. To determine if augmenting microglial activation would provide a potential therapeutic strategy, we administered granulocyte-macrophage colony-stimulating factor (GM-CSF) to WNV-infected mice. Recombinant human GM-CSF (rHuGMCSF) (sargramostim [Leukine]) is an FDA-approved drug used to increase white blood cells following leukopenia-inducing chemotherapy or bone marrow transplantation. Daily treatment of both uninfected and WNV-infected mice with subcutaneous injections of GM-CSF resulted in microglial proliferation and activation as indicated by the enhanced expression of the microglia activation marker ionized calcium binding adaptor molecule 1 (Iba1) and several microglia-associated inflammatory cytokines, including CCL2 (C-C motif chemokine ligand 2), interleukin 6 (IL-6), and IL-10. In addition, more microglia adopted an activated morphology as demonstrated by increased sizes and more pronounced processes. GM-CSF-induced microglial activation in WNV-infected mice was associated with reduced viral titers and apoptotic activity (caspase 3) in the brains of WNV-infected mice and significantly increased survival. WNV-infected ex vivo brain slice cultures (BSCs) treated with GM-CSF also showed reduced viral titers and caspase 3 apoptotic cell death, indicating that GM-CSF specifically targets the CNS and that its actions are not dependent on peripheral immune activity. Our studies suggest that stimulation of microglial activation may be a viable therapeutic approach for the treatment of WNV neuroinvasive disease. IMPORTANCE Although rare, WNV encephalitis poses a devastating health concern, with few treatment options and frequent long-term neurological sequelae. Currently, there are no human vaccines or specific antivirals against WNV infections, so further research into potential new therapeutic agents is critical. This study presents a novel treatment option for WNV infections using GM-CSF and lays the foundation for further studies into the use of GM-CSF as a treatment for WNV encephalitis as well as a potential treatment for other viral infections.
Collapse
Affiliation(s)
- Sarah Stonedahl
- Department of Immunology, University of Colorado, Aurora, Colorado, USA
- Department of Microbiology, University of Colorado, Aurora, Colorado, USA
| | - J. Smith Leser
- Department of Neurology, University of Colorado, Aurora, Colorado, USA
| | - Penny Clarke
- Department of Neurology, University of Colorado, Aurora, Colorado, USA
| | - Huntington Potter
- Department of Neurology, University of Colorado, Aurora, Colorado, USA
- University of Colorado Alzheimer’s and Cognition Center, Aurora, Colorado, USA
- Linda Crnic Institute for Down Syndrome, Aurora, Colorado, USA
| | - Timothy D. Boyd
- University of Colorado Alzheimer’s and Cognition Center, Aurora, Colorado, USA
- Linda Crnic Institute for Down Syndrome, Aurora, Colorado, USA
| | - Kenneth L. Tyler
- Department of Neurology, University of Colorado, Aurora, Colorado, USA
- Division of Infectious Disease, Department of Medicine, University of Colorado, Aurora, Colorado, USA
- Denver VA Medical Center, Aurora, Colorado, USA
| |
Collapse
|
5
|
Weber GE, Khrestian M, Tuason ED, Shao Y, Pillai J, Rao S, Feng H, Zhou Y, Cheng F, DeSilva TM, Stauffer S, Leverenz JB, Bekris LM. Peripheral sTREM2-Related Inflammatory Activity Alterations in Early-Stage Alzheimer's Disease. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2022; 208:2283-2299. [PMID: 35523454 PMCID: PMC9117433 DOI: 10.4049/jimmunol.2100771] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/05/2021] [Accepted: 03/07/2022] [Indexed: 05/17/2023]
Abstract
Alzheimer's disease (AD) has been linked to multiple immune system-related genetic variants. Triggering receptor expressed on myeloid cells 2 (TREM2) genetic variants are risk factors for AD and other neurodegenerative diseases. In addition, soluble TREM2 (sTREM2) isoform is elevated in cerebrospinal fluid in the early stages of AD and is associated with slower cognitive decline in a disease stage-dependent manner. Multiple studies have reported an altered peripheral immune response in AD. However, less is known about the relationship between peripheral sTREM2 and an altered peripheral immune response in AD. The objective of this study was to explore the relationship between human plasma sTREM2 and inflammatory activity in AD. The hypothesis of this exploratory study was that sTREM2-related inflammatory activity differs by AD stage. We observed different patterns of inflammatory activity across AD stages that implicate early-stage alterations in peripheral sTREM2-related inflammatory activity in AD. Notably, fractalkine showed a significant relationship with sTREM2 across different analyses in the control groups that was lost in later AD-related stages with high levels in mild cognitive impairment. Although multiple other inflammatory factors either differed significantly between groups or were significantly correlated with sTREM2 within specific groups, three inflammatory factors (fibroblast growth factor-2, GM-CSF, and IL-1β) are notable because they exhibited both lower levels in AD, compared with mild cognitive impairment, and a change in the relationship with sTREM2. This evidence provides important support to the hypothesis that sTREM2-related inflammatory activity alterations are AD stage specific and provides critical information for therapeutic strategies focused on the immune response.
Collapse
Affiliation(s)
- Grace E Weber
- Genomic Medicine Institute, Cleveland Clinic, Cleveland, OH
| | | | | | - Yvonne Shao
- Genomic Medicine Institute, Cleveland Clinic, Cleveland, OH
| | - Jagan Pillai
- Lou Ruvo Center for Brain Health, Neurological Institute, Cleveland Clinic, Cleveland, OH
| | - Stephen Rao
- Lou Ruvo Center for Brain Health, Neurological Institute, Cleveland Clinic, Cleveland, OH
| | - Hao Feng
- Department of Population and Quantitative Health Sciences, Case Western Reserve University School of Medicine, Cleveland, OH
| | - Yadi Zhou
- Genomic Medicine Institute, Cleveland Clinic, Cleveland, OH
| | - Feixiong Cheng
- Genomic Medicine Institute, Cleveland Clinic, Cleveland, OH
| | - Tara M DeSilva
- Department of Neurosciences, Cleveland Clinic, Cleveland, OH; and
| | - Shaun Stauffer
- Center for Therapeutics Discovery, Cleveland Clinic, Cleveland, OH
| | - James B Leverenz
- Lou Ruvo Center for Brain Health, Neurological Institute, Cleveland Clinic, Cleveland, OH
| | - Lynn M Bekris
- Genomic Medicine Institute, Cleveland Clinic, Cleveland, OH;
| |
Collapse
|
6
|
Kendall LV, Boyd TD, Sillau SH, Bosco-Lauth A, Markham N, Fong D, Clarke P, Tyler KL, Potter H. GM-CSF Promotes Immune Response and Survival in a Mouse Model of COVID-19. RESEARCH SQUARE 2022:rs.3.rs-1213395. [PMID: 35118463 PMCID: PMC8811947 DOI: 10.21203/rs.3.rs-1213395/v1] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 01/16/2023]
Abstract
COVID-19 results in increased expression of inflammatory cytokines, but inflammation-targeting clinical trials have yielded poor to mixed results. Our studies of other disorders with an inflammatory component, including Alzheimer's disease, chemobrain, Down syndrome, normal aging, and West Nile Virus infection, showed that treatment with the 'pro-inflammatory' cytokine granulocyte-macrophage colony stimulating factor (GM-CSF) in humans or mouse models alleviated clinical, behavioral, and pathological features. We proposed that human recombinant GM-CSF (sargramostim) be repurposed to promote both the innate and adaptive immune responses in COVID-19 to reduce viral load and mortality1. Here, we report the results of a placebo-controlled study of GM-CSF in human ACE2 transgenic mice inoculated intranasally with SARS-CoV2 virus, a model of COVID-19. Infection resulted in high viral titers in lungs and brains and over 85% mortality. GM-CSF treatment beginning one day after infection increased anti-viral antibody titers, lowered mean lung viral titers proportionately (p=0.0020) and increased the odds of long-term survival by up to 5.8-fold (p=0.0358), compared to placebo. These findings suggest that, as an activator of both the innate and adaptive immune systems, GM-CSF/sargramostim may be an effective COVID-19 therapy with the potential to protect from re-infection more effectively than treatment with antiviral drugs or monoclonal antibodies.
Collapse
Affiliation(s)
- L V Kendall
- Colorado State University, Department of Microbiology, Immunology and Pathology, Fort Collins, CO
| | - T D Boyd
- University of Colorado Alzheimer's and Cognition Center, Aurora, CO
- Linda Crnic Institute for Down Syndrome, University of Colorado School of Medicine, Aurora, CO
| | - S H Sillau
- University of Colorado Alzheimer's and Cognition Center, Aurora, CO
- Department of Neurology, University of Colorado School of Medicine, Aurora, CO
| | - A Bosco-Lauth
- Colorado State University, Department of Biomedical Sciences, Fort Collins, CO
| | - N Markham
- University of Colorado Alzheimer's and Cognition Center, Aurora, CO
- Linda Crnic Institute for Down Syndrome, University of Colorado School of Medicine, Aurora, CO
| | - D Fong
- Department of Pathology, University of Colorado Anschutz School of Medicine, Aurora, CO
| | - P Clarke
- Department of Neurology, University of Colorado School of Medicine, Aurora, CO
| | - K L Tyler
- Department of Neurology, University of Colorado School of Medicine, Aurora, CO
- Denver VA Medical Center, Denver CO
- Departments of Immunology and Microbiology, and Medicine, University of Colorado School of Medicine, Aurora, CO
| | - H Potter
- University of Colorado Alzheimer's and Cognition Center, Aurora, CO
- Linda Crnic Institute for Down Syndrome, University of Colorado School of Medicine, Aurora, CO
- Department of Neurology, University of Colorado School of Medicine, Aurora, CO
| |
Collapse
|
7
|
Golde TE. Disease-Modifying Therapies for Alzheimer's Disease: More Questions than Answers. Neurotherapeutics 2022; 19:209-227. [PMID: 35229269 PMCID: PMC8885119 DOI: 10.1007/s13311-022-01201-2] [Citation(s) in RCA: 66] [Impact Index Per Article: 22.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 02/09/2022] [Indexed: 12/17/2022] Open
Abstract
Scientific advances over the last four decades have steadily infused the Alzheimer's disease (AD) field with great optimism that therapies targeting Aβ, amyloid, tau, and innate immune activation states in the brain would provide disease modification. Unfortunately, this optimistic scenario has not yet played out. Though a recent approval of the anti-Aβ aggregate binding antibody, Aduhelm (aducanumab), as a "disease-modifying therapy for AD" is viewed by some as a breakthrough, many remain unconvinced by the data underlying this approval. Collectively, we have not succeeded in changing AD from a largely untreatable, inevitable, and incurable disease to a treatable, preventable, and curable one. Here, I will review the major foci of the AD "disease-modifying" therapeutic pipeline and some of the "open questions" that remain in terms of these therapeutic approaches. I will conclude the review by discussing how we, as a field, might adjust our approach, learning from our past failures to ensure future success.
Collapse
Affiliation(s)
- Todd E Golde
- Departments of Neuroscience and Neurology, Center for Translational Research in Neurodegenerative Disease, Evelyn F. and William L. McKnight Brain Institute, Norman Fixel Institute for Neurological Diseases, University of Florida, Gainesville, FL, USA.
| |
Collapse
|
8
|
Ahmed MM, Johnson NR, Boyd TD, Coughlan C, Chial HJ, Potter H. Innate Immune System Activation and Neuroinflammation in Down Syndrome and Neurodegeneration: Therapeutic Targets or Partners? Front Aging Neurosci 2021; 13:718426. [PMID: 34603007 PMCID: PMC8481947 DOI: 10.3389/fnagi.2021.718426] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2021] [Accepted: 08/16/2021] [Indexed: 12/24/2022] Open
Abstract
Innate immune system activation and inflammation are associated with and may contribute to clinical outcomes in people with Down syndrome (DS), neurodegenerative diseases such as Alzheimer's disease (AD), and normal aging. In addition to serving as potential diagnostic biomarkers, innate immune system activation and inflammation may play a contributing or causal role in these conditions, leading to the hypothesis that effective therapies should seek to dampen their effects. However, recent intervention studies with the innate immune system activator granulocyte-macrophage colony-stimulating factor (GM-CSF) in animal models of DS, AD, and normal aging, and in an AD clinical trial suggest that activating the innate immune system and inflammation may instead be therapeutic. We consider evidence that DS, AD, and normal aging are accompanied by innate immune system activation and inflammation and discuss whether and when during the disease process it may be therapeutically beneficial to suppress or promote such activation.
Collapse
Affiliation(s)
- Md. Mahiuddin Ahmed
- Department of Neurology, University of Colorado Anschutz Medical Campus, Aurora, CO, United States
- University of Colorado Alzheimer’s and Cognition Center, University of Colorado Anschutz Medical Campus, Aurora, CO, United States
- Linda Crnic Institute for Down Syndrome, University of Colorado Anschutz Medical Campus, Aurora, CO, United States
| | - Noah R. Johnson
- Department of Neurology, University of Colorado Anschutz Medical Campus, Aurora, CO, United States
- University of Colorado Alzheimer’s and Cognition Center, University of Colorado Anschutz Medical Campus, Aurora, CO, United States
- Linda Crnic Institute for Down Syndrome, University of Colorado Anschutz Medical Campus, Aurora, CO, United States
| | - Timothy D. Boyd
- Department of Neurology, University of Colorado Anschutz Medical Campus, Aurora, CO, United States
- University of Colorado Alzheimer’s and Cognition Center, University of Colorado Anschutz Medical Campus, Aurora, CO, United States
- Partner Therapeutics, Inc., Lexington, MA, United States
| | - Christina Coughlan
- Department of Neurology, University of Colorado Anschutz Medical Campus, Aurora, CO, United States
- University of Colorado Alzheimer’s and Cognition Center, University of Colorado Anschutz Medical Campus, Aurora, CO, United States
- Linda Crnic Institute for Down Syndrome, University of Colorado Anschutz Medical Campus, Aurora, CO, United States
| | - Heidi J. Chial
- Department of Neurology, University of Colorado Anschutz Medical Campus, Aurora, CO, United States
- University of Colorado Alzheimer’s and Cognition Center, University of Colorado Anschutz Medical Campus, Aurora, CO, United States
- Linda Crnic Institute for Down Syndrome, University of Colorado Anschutz Medical Campus, Aurora, CO, United States
| | - Huntington Potter
- Department of Neurology, University of Colorado Anschutz Medical Campus, Aurora, CO, United States
- University of Colorado Alzheimer’s and Cognition Center, University of Colorado Anschutz Medical Campus, Aurora, CO, United States
- Linda Crnic Institute for Down Syndrome, University of Colorado Anschutz Medical Campus, Aurora, CO, United States
| |
Collapse
|
9
|
Lazarus HM, Ragsdale CE, Gale RP, Lyman GH. Sargramostim (rhu GM-CSF) as Cancer Therapy (Systematic Review) and An Immunomodulator. A Drug Before Its Time? Front Immunol 2021; 12:706186. [PMID: 34484202 PMCID: PMC8416151 DOI: 10.3389/fimmu.2021.706186] [Citation(s) in RCA: 27] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2021] [Accepted: 07/26/2021] [Indexed: 12/31/2022] Open
Abstract
BACKGROUND Sargramostim [recombinant human granulocyte-macrophage colony-stimulating factor (rhu GM-CSF)] was approved by US FDA in 1991 to accelerate bone marrow recovery in diverse settings of bone marrow failure and is designated on the list of FDA Essential Medicines, Medical Countermeasures, and Critical Inputs. Other important biological activities including accelerating tissue repair and modulating host immunity to infection and cancer via the innate and adaptive immune systems are reported in pre-clinical models but incompletely studied in humans. OBJECTIVE Assess safety and efficacy of sargramostim in cancer and other diverse experimental and clinical settings. METHODS AND RESULTS We systematically reviewed PubMed, Cochrane and TRIP databases for clinical data on sargramostim in cancer. In a variety of settings, sargramostim after exposure to bone marrow-suppressing agents accelerated hematologic recovery resulting in fewer infections, less therapy-related toxicity and sometimes improved survival. As an immune modulator, sargramostim also enhanced anti-cancer responses in solid cancers when combined with conventional therapies, for example with immune checkpoint inhibitors and monoclonal antibodies. CONCLUSIONS Sargramostim accelerates hematologic recovery in diverse clinical settings and enhances anti-cancer responses with a favorable safety profile. Uses other than in hematologic recovery are less-well studied; more data are needed on immune-enhancing benefits. We envision significantly expanded use of sargramostim in varied immune settings. Sargramostim has the potential to reverse the immune suppression associated with sepsis, trauma, acute respiratory distress syndrome (ARDS) and COVID-19. Further, sargramostim therapy has been promising in the adjuvant setting with vaccines and for anti-microbial-resistant infections and treating autoimmune pulmonary alveolar proteinosis and gastrointestinal, peripheral arterial and neuro-inflammatory diseases. It also may be useful as an adjuvant in anti-cancer immunotherapy.
Collapse
Affiliation(s)
- Hillard M. Lazarus
- Department of Medicine, Case Western Reserve University, Cleveland, OH, United States
| | | | - Robert Peter Gale
- Centre for Haematology, Department of Immunology and Inflammation, Imperial College London, London, United Kingdom
| | - Gary H. Lyman
- Public Health Sciences and Clinical Research Divisions, Fred Hutchinson Cancer Research Center, Seattle, WA, United States
| |
Collapse
|
10
|
Potter H, Woodcock JH, Boyd TD, Coughlan CM, O'Shaughnessy JR, Borges MT, Thaker AA, Raj BA, Adamszuk K, Scott D, Adame V, Anton P, Chial HJ, Gray H, Daniels J, Stocker ME, Sillau SH. Safety and efficacy of sargramostim (GM-CSF) in the treatment of Alzheimer's disease. ALZHEIMER'S & DEMENTIA (NEW YORK, N. Y.) 2021; 7:e12158. [PMID: 33778150 PMCID: PMC7988877 DOI: 10.1002/trc2.12158] [Citation(s) in RCA: 40] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/20/2021] [Accepted: 02/05/2021] [Indexed: 02/06/2023]
Abstract
INTRODUCTION Inflammatory markers have long been observed in the brain, cerebrospinal fluid (CSF), and plasma of Alzheimer's disease (AD) patients, suggesting that inflammation contributes to AD and might be a therapeutic target. However, non-steroidal anti-inflammatory drug trials in AD and mild cognitive impairment (MCI) failed to show benefit. Our previous work seeking to understand why people with the inflammatory disease rheumatoid arthritis are protected from AD found that short-term treatment of transgenic AD mice with the pro-inflammatory cytokine granulocyte-macrophage colony-stimulating factor (GM-CSF) led to an increase in activated microglia, a 50% reduction in amyloid load, an increase in synaptic area, and improvement in spatial memory to normal. These results called into question the consensus view that inflammation is solely detrimental in AD. Here, we tested our hypothesis that modulation of the innate immune system might similarly be used to treat AD in humans by investigating the ability of GM-CSF/sargramostim to safely ameliorate AD symptoms/pathology. METHODS A randomized, double-blind, placebo-controlled trial was conducted in mild-to-moderate AD participants (NCT01409915). Treatments (20 participants/group) occurred 5 days/week for 3 weeks plus two follow-up (FU) visits (FU1 at 45 days and FU2 at 90 days) with neurological, neuropsychological, blood biomarker, and imaging assessments. RESULTS Sargramostim treatment expectedly changed innate immune system markers, with no drug-related serious adverse events or amyloid-related imaging abnormalities. At end of treatment (EOT), the Mini-Mental State Examination score of the sargramostim group increased compared to baseline (P = .0074) and compared to placebo (P = .0370); the treatment effect persisted at FU1 (P = .0272). Plasma markers of amyloid beta (Aβ40 [decreased in AD]) increased 10% (P = .0105); plasma markers of neurodegeneration (total tau and UCH-L1) decreased 24% (P = .0174) and 42% (P = .0019), respectively, after sargramostim treatment compared to placebo. DISCUSSION The innate immune system is a viable target for therapeutic intervention in AD. An extended treatment trial testing the long-term safety and efficacy of GM-CSF/sargramostim in AD is warranted.
Collapse
Affiliation(s)
- Huntington Potter
- Department of NeurologyUniversity of Colorado School of MedicineAuroraColoradoUSA
- University of Colorado Alzheimer's and Cognition CenterAuroraColoradoUSA
- Linda Crnic Institute for Down SyndromeUniversity of Colorado School of MedicineAuroraColoradoUSA
| | - Jonathan H. Woodcock
- Department of NeurologyUniversity of Colorado School of MedicineAuroraColoradoUSA
- University of Colorado Alzheimer's and Cognition CenterAuroraColoradoUSA
| | - Timothy D. Boyd
- University of Colorado Alzheimer's and Cognition CenterAuroraColoradoUSA
- Linda Crnic Institute for Down SyndromeUniversity of Colorado School of MedicineAuroraColoradoUSA
| | - Christina M. Coughlan
- Department of NeurologyUniversity of Colorado School of MedicineAuroraColoradoUSA
- University of Colorado Alzheimer's and Cognition CenterAuroraColoradoUSA
- Linda Crnic Institute for Down SyndromeUniversity of Colorado School of MedicineAuroraColoradoUSA
| | - John R. O'Shaughnessy
- Department of NeurologyUniversity of Colorado School of MedicineAuroraColoradoUSA
- University of Colorado Alzheimer's and Cognition CenterAuroraColoradoUSA
| | - Manuel T. Borges
- Department of NeurologyUniversity of Colorado School of MedicineAuroraColoradoUSA
- Department of RadiologyUniversity of Colorado School of MedicineAuroraColoradoUSA
| | - Ashesh A. Thaker
- Department of NeurologyUniversity of Colorado School of MedicineAuroraColoradoUSA
- Department of RadiologyUniversity of Colorado School of MedicineAuroraColoradoUSA
| | | | | | | | - Vanesa Adame
- University of Colorado Alzheimer's and Cognition CenterAuroraColoradoUSA
- Linda Crnic Institute for Down SyndromeUniversity of Colorado School of MedicineAuroraColoradoUSA
| | - Paige Anton
- University of Colorado Alzheimer's and Cognition CenterAuroraColoradoUSA
- Linda Crnic Institute for Down SyndromeUniversity of Colorado School of MedicineAuroraColoradoUSA
| | - Heidi J. Chial
- Department of NeurologyUniversity of Colorado School of MedicineAuroraColoradoUSA
- University of Colorado Alzheimer's and Cognition CenterAuroraColoradoUSA
- Linda Crnic Institute for Down SyndromeUniversity of Colorado School of MedicineAuroraColoradoUSA
| | - Helen Gray
- Department of NeurologyUniversity of Colorado School of MedicineAuroraColoradoUSA
- University of Colorado Alzheimer's and Cognition CenterAuroraColoradoUSA
| | - Joseph Daniels
- Department of NeurologyUniversity of Colorado School of MedicineAuroraColoradoUSA
- University of Colorado Alzheimer's and Cognition CenterAuroraColoradoUSA
| | - Michelle E. Stocker
- Department of NeurologyUniversity of Colorado School of MedicineAuroraColoradoUSA
- University of Colorado Alzheimer's and Cognition CenterAuroraColoradoUSA
| | - Stefan H. Sillau
- Department of NeurologyUniversity of Colorado School of MedicineAuroraColoradoUSA
- University of Colorado Alzheimer's and Cognition CenterAuroraColoradoUSA
| |
Collapse
|
11
|
Stonedahl S, Clarke P, Tyler KL. The Role of Microglia during West Nile Virus Infection of the Central Nervous System. Vaccines (Basel) 2020; 8:E485. [PMID: 32872152 PMCID: PMC7563127 DOI: 10.3390/vaccines8030485] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2020] [Revised: 08/22/2020] [Accepted: 08/26/2020] [Indexed: 12/28/2022] Open
Abstract
Encephalitis resulting from viral infections is a major cause of hospitalization and death worldwide. West Nile Virus (WNV) is a substantial health concern as it is one of the leading causes of viral encephalitis in the United States today. WNV infiltrates the central nervous system (CNS), where it directly infects neurons and induces neuronal cell death, in part, via activation of caspase 3-mediated apoptosis. WNV infection also induces neuroinflammation characterized by activation of innate immune cells, including microglia and astrocytes, production of inflammatory cytokines, breakdown of the blood-brain barrier, and infiltration of peripheral leukocytes. Microglia are the resident immune cells of the brain and monitor the CNS for signs of injury or pathogens. Following infection with WNV, microglia exhibit a change in morphology consistent with activation and are associated with increased expression of proinflammatory cytokines. Recent research has focused on deciphering the role of microglia during WNV encephalitis. Microglia play a protective role during infections by limiting viral growth and reducing mortality in mice. However, it also appears that activated microglia are triggered by T cells to mediate synaptic elimination at late times during infection, which may contribute to long-term neurological deficits following a neuroinvasive WNV infection. This review will discuss the important role of microglia in the pathogenesis of a neuroinvasive WNV infection. Knowledge of the precise role of microglia during a WNV infection may lead to a greater ability to treat and manage WNV encephalitis.
Collapse
Affiliation(s)
- Sarah Stonedahl
- Department of Immunology and Microbiology University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA;
| | - Penny Clarke
- Department of Neurology, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA
| | - Kenneth L. Tyler
- Department of Immunology and Microbiology, Infectious Disease, Medicine and Neurology, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA
- Department of Veterans Affairs, Aurora, CO 80045, USA
| |
Collapse
|
12
|
Potter H, Boyd TD, Clarke P, Pelak VS, Tyler KL. Recruiting the innate immune system with GM-CSF to fight viral diseases, including West Nile Virus encephalitis and COVID-19. F1000Res 2020; 9:345. [PMID: 32704352 PMCID: PMC7359749 DOI: 10.12688/f1000research.23729.1] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 04/30/2020] [Indexed: 01/08/2023] Open
Abstract
As the coronavirus disease 2019 (COVID-19) pandemic grows throughout the world, it is imperative that all approaches to ameliorating its effects be investigated, including repurposing drugs that show promise in other diseases. We have been investigating an approach to multiple disorders that involves recruiting the innate immune system to aid the body's healing and regenerative mechanism(s). In the case of West Nile Virus encephalitis and potentially COVID-19, the proposed intervention to stimulate the innate immune system may give the adaptive immune response the necessary time to develop, finish clearing the virus, and provide future immunity. Furthermore, we have found that GM-CSF-induced recruitment of the innate immune system is also able to reverse brain pathology, neuroinflammation and cognitive deficits in mouse models of Alzheimer's disease and Down syndrome, as well as improving cognition in normal aging and in human patients with cognitive deficits due to chemotherapy, both of which exhibit neuroinflammation. Others have shown that GM-CSF is an effective treatment for both bacterial and viral pneumonias, and their associated inflammation, in animals and that it has successfully treated pneumonia-associated Acute Respiratory Distress Syndrome in humans. These and other data strongly suggest that GM-CSF may be an effective treatment for many viral infections, including COVID-19.
Collapse
Affiliation(s)
- Huntington Potter
- Department of Neurology, University of Colorado Anschutz Medical Campus, Aurora, CO, 80045, USA
- University of Colorado Alzheimer's and Cognition Center, Aurora, CO, 80045, USA
- Linda Crnic Institute for Down Syndrome, Aurora, CO, 80045, USA
| | - Timothy D. Boyd
- University of Colorado Alzheimer's and Cognition Center, Aurora, CO, 80045, USA
- Linda Crnic Institute for Down Syndrome, Aurora, CO, 80045, USA
| | - Penny Clarke
- Department of Neurology, University of Colorado Anschutz Medical Campus, Aurora, CO, 80045, USA
| | - Victoria S. Pelak
- Department of Neurology, University of Colorado Anschutz Medical Campus, Aurora, CO, 80045, USA
- University of Colorado Alzheimer's and Cognition Center, Aurora, CO, 80045, USA
| | - Kenneth L. Tyler
- Department of Neurology, University of Colorado Anschutz Medical Campus, Aurora, CO, 80045, USA
| |
Collapse
|
13
|
Cao W, Zheng H. Peripheral immune system in aging and Alzheimer's disease. Mol Neurodegener 2018; 13:51. [PMID: 30285785 PMCID: PMC6169078 DOI: 10.1186/s13024-018-0284-2] [Citation(s) in RCA: 143] [Impact Index Per Article: 20.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2018] [Accepted: 09/21/2018] [Indexed: 12/13/2022] Open
Abstract
Alzheimer's disease (AD) represents an urgent public health mandate. AD is no longer considered a neural-centric disease; rather, a plethora of recent studies strongly implicate a critical role played by neuroinflammation in the pathogeneses of AD and other neurodegenerative conditions. A close functional connection between the immune system and central nervous system is increasingly recognized. In late-onset AD, aging represents the most significant risk factor. Here, from an immunological perspective, we summarize the prominent molecular and cellular changes in the periphery of aging individuals and AD patients. Moreover, we review the knowledge gained in the past several years that implicate specific arms of the peripheral immune system and other types of immune responses in modulating AD progression. Taken together, these findings collectively emphasize a dynamic role of a concert of brain-extrinsic, peripheral signals in the aging and degenerative processes in the CNS. We believe that a systematic view synthesizing the vast amounts of existing results will help guide the development of next-generation therapeutics and inform future directions of AD investigation.
Collapse
Affiliation(s)
- Wei Cao
- Department of Molecular and Human Genetics, Baylor College of Medicine, Huffington Center on Aging, Houston, TX, 77030, USA.
| | - Hui Zheng
- Department of Molecular and Human Genetics, Baylor College of Medicine, Huffington Center on Aging, Houston, TX, 77030, USA.
| |
Collapse
|
14
|
Hamlett ED, Ledreux A, Potter H, Chial HJ, Patterson D, Espinosa JM, Bettcher BM, Granholm AC. Exosomal biomarkers in Down syndrome and Alzheimer's disease. Free Radic Biol Med 2018; 114:110-121. [PMID: 28882786 PMCID: PMC6135098 DOI: 10.1016/j.freeradbiomed.2017.08.028] [Citation(s) in RCA: 60] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/06/2017] [Revised: 08/30/2017] [Accepted: 08/31/2017] [Indexed: 02/07/2023]
Abstract
Every person with Down syndrome (DS) has the characteristic features of Alzheimer's disease (AD) neuropathology in their brain by the age of forty, and most go on to develop AD dementia. Since people with DS show highly variable levels of baseline function, it is often difficult to identify early signs of dementia in this population. The discovery of blood biomarkers predictive of dementia onset and/or progression in DS is critical for developing effective clinical diagnostics. Our recent studies show that neuron-derived exosomes, which are small extracellular vesicles secreted by most cells in the body, contain elevated levels of amyloid-beta peptides and phosphorylated-Tau that could indicate a preclinical AD phase in people with DS starting in childhood. We also found that the relative levels of these biomarkers were altered following dementia onset. Exosome release and signaling are dependent on cellular redox homeostasis as well as on inflammatory processes, and exosomes may be involved in the immune response, suggesting a dual role as both triggers of inflammation in the brain and propagators of inflammatory signals between brain regions. Based on recently reported connections between inflammatory processes and exosome release, the elevated neuroinflammatory state observed in people with DS may affect exosomal AD biomarkers. Herein, we discuss findings from studies of people with DS, people with DS and AD (DS-AD), and mouse models of DS showing new connections between neuroinflammatory pathways, oxidative stress, exosomes, and exosome-mediated signaling, which may inform future AD diagnostics, preventions, and treatments in the DS population as well as in the general population.
Collapse
Affiliation(s)
- Eric D Hamlett
- Knoebel Institute for Healthy Aging and the Department of Biological Sciences, University of Denver, Denver, CO, USA; Medical University of South Carolina, Charleston, SC, USA
| | - Aurélie Ledreux
- Knoebel Institute for Healthy Aging and the Department of Biological Sciences, University of Denver, Denver, CO, USA
| | - Huntington Potter
- Rocky Mountain Alzheimer's Disease Center, University of Colorado Anschutz Medical Campus, Denver, CO, USA; Linda Crnic Institute for Down Syndrome, University of Colorado Anschutz Medical Campus, Denver, CO, USA; Department of Neurology, University of Colorado Anschutz Medical Campus, Denver, CO, USA
| | - Heidi J Chial
- Rocky Mountain Alzheimer's Disease Center, University of Colorado Anschutz Medical Campus, Denver, CO, USA; Linda Crnic Institute for Down Syndrome, University of Colorado Anschutz Medical Campus, Denver, CO, USA; Department of Neurology, University of Colorado Anschutz Medical Campus, Denver, CO, USA
| | - David Patterson
- Knoebel Institute for Healthy Aging and the Department of Biological Sciences, University of Denver, Denver, CO, USA
| | - Joaquin M Espinosa
- Linda Crnic Institute for Down Syndrome, University of Colorado Anschutz Medical Campus, Denver, CO, USA; Department of Pharmacology, University of Colorado Anschutz Medical Campus, Denver, CO, USA
| | - Brianne M Bettcher
- Rocky Mountain Alzheimer's Disease Center, University of Colorado Anschutz Medical Campus, Denver, CO, USA; Linda Crnic Institute for Down Syndrome, University of Colorado Anschutz Medical Campus, Denver, CO, USA; Department of Neurology, University of Colorado Anschutz Medical Campus, Denver, CO, USA; Department of Neurosurgery, University of Colorado Anschutz Medical Campus, Denver, CO, USA
| | - Ann-Charlotte Granholm
- Knoebel Institute for Healthy Aging and the Department of Biological Sciences, University of Denver, Denver, CO, USA; Medical University of South Carolina, Charleston, SC, USA.
| |
Collapse
|
15
|
Comi C, Tondo G. Insights into the protective role of immunity in neurodegenerative disease. Neural Regen Res 2017; 12:64-65. [PMID: 28250745 PMCID: PMC5319239 DOI: 10.4103/1673-5374.198980] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/04/2022] Open
Affiliation(s)
- Cristoforo Comi
- Neurology Unit, Department of Translational Medicine, University of Piemonte Orientale, Novara, Italy; Interdisciplinary Research Center of Autoimmune Diseases (IRCAD), University of Piemonte Orientale, Novara, Italy
| | - Giacomo Tondo
- Neurology Unit, Department of Translational Medicine, University of Piemonte Orientale, Novara, Italy
| |
Collapse
|
16
|
Shanmugalingam U, Jadavji NM, Smith PD. Role of granulocyte macrophage colony stimulating factor in regeneration of the central nervous system. Neural Regen Res 2016; 11:902-3. [PMID: 27482209 PMCID: PMC4962578 DOI: 10.4103/1673-5374.184479] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Affiliation(s)
| | - Nafisa M Jadavji
- Department of Neuroscience, Carleton University, Ottawa, ON, Canada
| | - Patrice D Smith
- Department of Neuroscience, Carleton University, Ottawa, ON, Canada
| |
Collapse
|
17
|
Heinzelman P, Schoborg JA, Jewett MC. pH responsive granulocyte colony-stimulating factor variants with implications for treating Alzheimer's disease and other central nervous system disorders. Protein Eng Des Sel 2015; 28:481-9. [PMID: 25877663 PMCID: PMC4596278 DOI: 10.1093/protein/gzv022] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2014] [Revised: 02/27/2015] [Accepted: 03/06/2015] [Indexed: 12/14/2022] Open
Abstract
Systemic injection of granulocyte colony-stimulating factor (G-CSF) has yielded encouraging results in treating Alzheimer's Disease (AD) and other central nervous system (CNS) disorders. Making G-CSF a viable AD therapeutic will, however, require increasing G-CSF's ability to stimulate neurons within the brain. This objective could be realized by increasing transcytosis of G-CSF across the blood brain barrier (BBB). An established correlation between G-CSF receptor (G-CSFR) binding pH responsiveness and increased recycling of G-CSF to the cell exterior after endocytosis motivated development of G-CSF variants with highly pH responsive G-CSFR binding affinities. These variants will be used in future validation of our hypothesis that increased BBB transcytosis can enhance G-CSF therapeutic efficacy. Flow cytometric screening of a yeast-displayed library in which G-CSF/G-CSFR interface residues were mutated to histidine yielded a G-CSF triple His mutant (L109H/D110H/Q120H) with highly pH responsive binding affinity. This variant's KD, measured by surface plasmon resonance (SPR), increases ∼20-fold as pH decreases from 7.4 to below histidine's pKa of ∼6.0; an increase 2-fold greater than for previously reported G-CSF His mutants. Cell-free protein synthesis (CFPS) enabled expression and purification of soluble, bioactive G-CSF triple His variant protein, an outcome inaccessible via Escherichia coli inclusion body refolding. This purification and bioactivity validation will enable future identification of correlations between pH responsiveness and transcytosis in BBB cell culture model and animal experiments. Furthermore, the library screening and CFPS methods employed here could be applied to developing other pH responsive hematopoietic or neurotrophic factors for treating CNS disorders.
Collapse
Affiliation(s)
- Pete Heinzelman
- Department of Chemical, Biological and Materials Engineering, University of Oklahoma, Sarkeys Energy Center, 100 East Boyd Street, Room T-301, Norman, OK 73019, USA
| | - Jennifer A Schoborg
- Department of Chemical and Biological Engineering, Northwestern University, Evanston, IL 60208-3120, USA
| | - Michael C Jewett
- Department of Chemical and Biological Engineering, Northwestern University, Evanston, IL 60208-3120, USA
| |
Collapse
|