Go to JCI Insight
  • About
  • Editors
  • Consulting Editors
  • For authors
  • Publication ethics
  • Publication alerts by email
  • Advertising
  • Job board
  • Contact
  • Clinical Research and Public Health
  • Current issue
  • Past issues
  • By specialty
    • COVID-19
    • Cardiology
    • Gastroenterology
    • Immunology
    • Metabolism
    • Nephrology
    • Neuroscience
    • Oncology
    • Pulmonology
    • Vascular biology
    • All ...
  • Videos
    • Conversations with Giants in Medicine
    • Video Abstracts
  • Reviews
    • View all reviews ...
    • Complement Biology and Therapeutics (May 2025)
    • Evolving insights into MASLD and MASH pathogenesis and treatment (Apr 2025)
    • Microbiome in Health and Disease (Feb 2025)
    • Substance Use Disorders (Oct 2024)
    • Clonal Hematopoiesis (Oct 2024)
    • Sex Differences in Medicine (Sep 2024)
    • Vascular Malformations (Apr 2024)
    • View all review series ...
  • Viewpoint
  • Collections
    • In-Press Preview
    • Clinical Research and Public Health
    • Research Letters
    • Letters to the Editor
    • Editorials
    • Commentaries
    • Editor's notes
    • Reviews
    • Viewpoints
    • 100th anniversary
    • Top read articles

  • Current issue
  • Past issues
  • Specialties
  • Reviews
  • Review series
  • Conversations with Giants in Medicine
  • Video Abstracts
  • In-Press Preview
  • Clinical Research and Public Health
  • Research Letters
  • Letters to the Editor
  • Editorials
  • Commentaries
  • Editor's notes
  • Reviews
  • Viewpoints
  • 100th anniversary
  • Top read articles
  • About
  • Editors
  • Consulting Editors
  • For authors
  • Publication ethics
  • Publication alerts by email
  • Advertising
  • Job board
  • Contact

Clinical Research and Public Health

  • 387 Articles
  • 0 Posts
  • ← Previous
  • 1
  • 2
  • …
  • 22
  • 23
  • 24
  • …
  • 38
  • 39
  • Next →
COVID-19 survival associates with the immunoglobulin response to the SARS-CoV-2 spike Receptor Binding Domain
Massimiliano Secchi, … , Lorenzo Piemonti, Vito Lampasona
Massimiliano Secchi, … , Lorenzo Piemonti, Vito Lampasona
Published September 29, 2020
Citation Information: J Clin Invest. 2020. https://doi.org/10.1172/JCI142804.
View: Text | PDF

COVID-19 survival associates with the immunoglobulin response to the SARS-CoV-2 spike Receptor Binding Domain

  • Text
  • PDF
Abstract

BACKGROUND. Serological assays are of critical importance to investigate correlates of response and protection in COVID-19, to define previous exposure to SARS-CoV-2 in populations and to verify the development of an adaptive immune response in infected individuals. METHODS. We studied 509 confirmed COVID-19 patients from the San Raffaele Hospital of Milan and 480 pre-pandemic organ donor sera collected in 2010-2012. Using fluid-phase luciferase immune precipitation (LIPS) assays, we characterized IgG, IgM, IgA antibodies to the spike Receptor Binding Domain (RBD), S1+S2, nucleocapsid, and ORF6 to 10 of SARS-CoV-2, to the HCoV-OC43 and HCoV-HKU1 betacoronaviruses spike S2, and the H1N1Ca2009 flu virus hemagglutinin. Sequential samples at 1 and 3 months post-hospital discharge were also tested in 95 patients for SARS-CoV-2 RBD antibodies. RESULTS. Antibodies developed rapidly against multiple SARS-CoV-2 antigens in 95% of patients by 4 weeks post-symptoms onset and IgG to the RBD increased until the 3rd month of follow-up. We observed a major synchronous expansion of antibodies to the HCoV-OC43 and HCoV-HKU1 spike S2. A likely co-infection with influenza was neither linked to a more severe presentation of the disease nor to a worse outcome. Of the measured antibody responses positivity for IgG against the SARS-CoV-2 spike RBD was predictive of survival. CONCLUSIONS. The measurement of antibodies to selected epitopes of SARS-CoV-2 antigens can offer a more accurate assessment of the humoral response in patients and its impact on survival. The presence of partially cross-reactive antibodies with other betacoronoviruses is likely to impact on serological assay specificity and interpretation.

Authors

Massimiliano Secchi, Elena Bazzigaluppi, Cristina Brigatti, Ilaria Marzinotto, Cristina Tresoldi, Patrizia Rovere-Querini, Andrea Poli, Antonella Castagna, Gabriella Scarlatti, Alberto Zangrillo, Fabio Ciceri, Lorenzo Piemonti, Vito Lampasona

×

Tobacco smoking induces metabolic reprogramming of renal cell carcinoma
James Reigle, … , Jarek Meller, Maria F. Czyzyk-Krzeska
James Reigle, … , Jarek Meller, Maria F. Czyzyk-Krzeska
Published September 24, 2020
Citation Information: J Clin Invest. 2020. https://doi.org/10.1172/JCI140522.
View: Text | PDF

Tobacco smoking induces metabolic reprogramming of renal cell carcinoma

  • Text
  • PDF
Abstract

Background. Clear cell renal cell carcinoma (ccRCC) is the most common histologically defined renal cancer. However, it is not a uniform disease and includes several genetic subtypes with different prognosis. ccRCC is also characterized by distinguished metabolic reprogramming. Tobacco smoking (TS) is an established risk factor for ccRCC with unknown effects on tumor pathobiology. Methods. We investigated the landscape of ccRCCs and paired normal kidney tissues (NKTs) using integrated transcriptomic, metabolomic and metallomic approaches in a cohort of never smokers (NS) and long-term current smokers (LTS) Caucasian males. Results. All three Omics domains consistentl identified a distinct metabolic subtype of ccRCCs in LTS, characterized by activation of oxidative phosphorylation (OxPhos) coupled with reprogramming of the malate-aspartate shuttle and metabolism of aspartate, glutamate, glutamine and histidine. Cadmium, copper and inorganic arsenic accumulated in LTS tumors showing redistribution among intracellular pools, including relocation of copper into the cytochrome c oxidase complex. Gene expression signature based on the LTS metabolic subtype provided prognostic stratification of The Cancer Genome Atlas (TCGA) ccRCC tumors that was independent from genomic alterations. Conclusions. The work identifies the TS related metabolic subtype of ccRCC with vulnerabilities that can be exploited for precision medicine approaches targeting metabolic pathways. The results provide rationale for the development of metabolic biomarkers with diagnostic and prognostic applications using evaluation of OxPhos status. The metallomic analysis reveals the role of disrupted metal homeostasis in ccRCC highlighting the importance of studying effects of metals from e-cigarettes and environmental exposures.

Authors

James Reigle, Dina Secic, Jacek Biesiada, Collin Wetzel, Behrouz Shamsaei, Johnson Chu, Yuanwei Zang, Xiang Zhang, Nicholas J. Talbot, Megan E. Bischoff, Yongzhen Zhang, Charuhas V. Thakar, Krishnanath Gaitonde, Abhinav Sidana, Hai Bui, John T. Cunningham, Qing Zhang, Laura S. Schmidt, W. Marston Linehan, Mario Medvedovic, David R. Plas, Julio A. Landero Figueroa, Jarek Meller, Maria F. Czyzyk-Krzeska

×

Cytomegalovirus viral load kinetics as surrogate endpoints after allogeneic transplantation
Elizabeth R. Duke, … , Joshua T. Schiffer, Michael Boeckh
Elizabeth R. Duke, … , Joshua T. Schiffer, Michael Boeckh
Published September 24, 2020
Citation Information: J Clin Invest. 2020. https://doi.org/10.1172/JCI133960.
View: Text | PDF

Cytomegalovirus viral load kinetics as surrogate endpoints after allogeneic transplantation

  • Text
  • PDF
Abstract

Background: Viral load surrogate endpoints transformed development of HIV and hepatitis C therapeutics. Surrogate endpoints for cytomegalovirus (CMV)-related morbidity and mortality could advance development of antiviral treatments. While observational data support using CMV viral load (VL) as a trial endpoint, randomized controlled trials (RCT) demonstrating direct associations between virologic markers and clinical endpoints are lacking. Methods: We performed CMV DNA polymerase chain reaction (PCR) on frozen serum samples from the only placebo-controlled RCT of ganciclovir for early treatment of CMV after hematopoietic cell transplantation (HCT). We used established criteria to assess VL kinetics as surrogates for CMV disease or death by weeks 8, 24, and 48 after randomization and quantified antiviral effects captured by each marker. We used ensemble-based machine learning to assess the predictive ability of VL kinetics and performed this analysis on a ganciclovir prophylaxis RCT for validation. Results: VL suppression with ganciclovir reduced cumulative incidence of CMV disease and death for 20 years after HCT. Mean VL, peak VL, and change in VL during the first five weeks of treatment fulfilled the Prentice definition for surrogacy, capturing > 95% of ganciclovir’s effect, and yielded highly sensitive and specific predictions by week 48. In the prophylaxis trial, viral shedding rate satisfied the Prentice definition for CMV disease by week 24. Conclusion: Our results support using CMV VL kinetics as surrogates for CMV disease, provide a framework for developing CMV preventative and therapeutic agents, and support reductions in viral load as the mechanism through which antivirals reduce CMV disease.

Authors

Elizabeth R. Duke, Brian D. Williamson, Bhavesh Borate, Jonathan L. Golob, Chiara Wychera, Terry Stevens-Ayers, Meei-Li Huang, Nicole Cossrow, Hong Wan, T. Christopher Mast, Morgan A. Marks, Mary Flowers, Keith R. Jerome, Lawrence Corey, Peter B. Gilbert, Joshua T. Schiffer, Michael Boeckh

×

Healthy donor T-cell responses to common cold coronaviruses and SARS-CoV-2
Bezawit A. Woldemeskel, … , Stuart C. Ray, Joel N. Blankson
Bezawit A. Woldemeskel, … , Stuart C. Ray, Joel N. Blankson
Published September 23, 2020
Citation Information: J Clin Invest. 2020. https://doi.org/10.1172/JCI143120.
View: Text | PDF

Healthy donor T-cell responses to common cold coronaviruses and SARS-CoV-2

  • Text
  • PDF
Abstract

BACKGROUND. The T cell responses to the common cold coronaviruses have not been well characterized. Pre-existing T cell immunity to SARS-CoV-2 has been reported, and a recent study suggested that this was due to cross-recognition of the novel coronavirus by T cells specific for the common cold coronaviruses. METHODS. We used the ELISpot assay to characterize the T cell responses against peptide pools derived from the spike protein of three common cold coronaviruses (HCoV-229E, HCoV-NL63, and HCoV-OC43) and SARS-CoV-2 in 21 healthy donors who were seronegative for SARS-CoV-2 and had no known exposure to the virus. An in vitro expansion culture assay was also used to analyze memory T cell responses. RESULTS. We found responses to the spike protein of the three common cold coronaviruses in many donors. We then focused on HCoV-NL63 and demonstrated broad T cell responses to the spike protein and identified 22 targeted peptides. Interestingly, only one subject had a significant response to SARS-CoV-2 spike or nucleocapsid protein in the ELISpot assay. In vitro expansion studies suggested that T cells specific for the HCoV-NL63 spike protein in this subject could also recognize SARS-CoV-2 spike protein peptide pools. CONCLUSIONS. Healthy donors have circulating T cells specific for the spike proteins of HCoV-NL63, HCoV-229E, and HCoV-OC43. T cell responses to SARS-CoV-2 spike and nucleocapsid proteins were present in only one subject and were potentially the result of cross-recognition by T cells specific for the common cold coronaviruses. Further studies are needed to determine whether this influences COVID-19 outcomes.

Authors

Bezawit A. Woldemeskel, Abena K. Kwaa, Caroline C. Garliss, Oliver Laeyendecker, Stuart C. Ray, Joel N. Blankson

×

Haploinsufficiency of immune checkpoint receptor CTLA4 induces a distinct neuroinflammatory disorder
Matthew K. Schindler, … , Daniel S. Reich, Gulbu Uzel
Matthew K. Schindler, … , Daniel S. Reich, Gulbu Uzel
Published September 21, 2020
Citation Information: J Clin Invest. 2020. https://doi.org/10.1172/JCI135947.
View: Text | PDF

Haploinsufficiency of immune checkpoint receptor CTLA4 induces a distinct neuroinflammatory disorder

  • Text
  • PDF
Abstract

BACKGROUND Cytotoxic T lymphocyte antigen 4 (CTLA4) is essential for immune homeostasis. Genetic mutations causing haploinsufficiency (CTLA4h) lead to a phenotypically heterogenous, immune-mediated disease that can include neuroinflammation. The neurological manifestations of CTLA4h are poorly characterized.METHODS We performed an observational natural history study of 50 patients with CTLA4h who were followed at the NIH. We analyzed clinical, radiological, immunological, and histopathological data.RESULTS Evidence for neuroinflammation was observed in 32% (n = 16 of 50) of patients in this cohort by magnetic resonance imaging (MRI) and/or by cerebrospinal fluid analysis. Clinical symptoms were commonly absent or mild in severity, with headaches as the leading complaint (n = 13 of 16). The most striking findings were relapsing, large, contrast-enhancing focal lesions in the brain and spinal cord observed on MRI. We detected inflammation in the cerebrospinal fluid and leptomeninges before the parenchyma. Brain biopsies of inflammatory lesions from 10 patients showed perivascular and intraparenchymal mixed cellular infiltrates with little accompanying demyelination or neuronal injury.CONCLUSIONS Neuroinflammation due to CTLA4h is mediated primarily by an infiltrative process with a distinct and striking dissociation between clinical symptoms and radiological findings in the majority of patients.FUNDING NIAID, NIH, Division of Intramural Research, NINDS, NIH, Division of Intramural Research, and the National Multiple Sclerosis Society–American Brain Foundation.TRIAL REGISTRATION ClinicalTrials.gov NCT00001355.

Authors

Matthew K. Schindler, Stefania Pittaluga, Yoshimi Enose-Akahata, Helen C. Su, V. Koneti Rao, Amy Rump, Steven Jacobson, Irene Cortese, Daniel S. Reich, Gulbu Uzel

×

The ABO Histo-Blood Group, endothelial activation, and acute respiratory distress syndrome risk in critical illness
John P. Reilly, … , Muredach P. Reilly, Jason D. Christie
John P. Reilly, … , Muredach P. Reilly, Jason D. Christie
Published September 15, 2020
Citation Information: J Clin Invest. 2020. https://doi.org/10.1172/JCI139700.
View: Text | PDF

The ABO Histo-Blood Group, endothelial activation, and acute respiratory distress syndrome risk in critical illness

  • Text
  • PDF
Abstract

BACKGROUND. The ABO histo-blood group is defined by carbohydrate modifications and is associated with risk for multiple diseases including Acute Respiratory Distress Syndrome (ARDS). We hypothesized that genetically determined blood subtype A1 is associated with increased risk of ARDS and markers of microvascular dysfunction and coagulation. METHODS. We conducted analyses in three cohorts of critically ill trauma and sepsis patients (n = 3,710) genotyped on genome-wide platforms to determine the association of the A1 blood type genotype with ARDS risk. We subsequently determined if associations were present in FUT2 defined non-secretors who lack ABO antigens on epithelium, but not endothelium. In a patient subgroup, we determined the associations of blood type with plasma levels of endothelial glycoproteins and disseminated intravascular coagulation (DIC). Lastly, we tested if blood type A was associated with less donor lung injury recovery during human ex vivo lung perfusion (EVLP). RESULTS. The A1 genotype was associated with a higher risk of moderate to severe ARDS relative to type O in all three populations. In sepsis, this relationship was strongest in non-pulmonary infections. The association persisted in non-secretors, suggesting a vascular mechanism. The A1 genotype was also associated with higher DIC risk as well as concentrations of thrombomodulin and von Willebrand Factor, which in turn were associated with ARDS risk. Blood type A was also associated with less lung injury recovery during EVLP. CONCLUSIONS. We identified a replicable association between ABO blood type A1 and risk of ARDS among the critically ill possibly mediated through microvascular dysfunction and coagulation.

Authors

John P. Reilly, Nuala J. Meyer, Michael G.S. Shashaty, Brian J. Anderson, Caroline Ittner, Thomas G. Dunn, Brian Lim, Caitlin Forker, Michael P. Bonk, Ethan D. Kotloff, Rui Feng, Edward Cantu, Nilam S. Mangalmurti, Carolyn S. Calfee, Michael A. Matthay, Carmen Mikacenic, Keith R. Walley, James A. Russell, David C. Christiani, Mark M. Wurfel, Paul N. Lanken, Muredach P. Reilly, Jason D. Christie

×

Favorable outcomes of COVID-19 in recipients of hematopoietic cell transplantation
Gunjan L. Shah, … , Genovefa A. Papanicolaou, Miguel-Angel Perales
Gunjan L. Shah, … , Genovefa A. Papanicolaou, Miguel-Angel Perales
Published September 8, 2020
Citation Information: J Clin Invest. 2020. https://doi.org/10.1172/JCI141777.
View: Text | PDF

Favorable outcomes of COVID-19 in recipients of hematopoietic cell transplantation

  • Text
  • PDF
Abstract

BACKGROUND. Understanding outcomes and immunologic characteristics of cellular therapy recipients with SARS-CoV-2 is critical to performing these potentially life-saving therapies in the COVID-19 era. In this study of recipients of allogeneic (Allo) and autologous (Auto) hematopoietic cell transplant and CD19-directed chimeric antigen receptor T cell therapy (CAR-T) at Memorial Sloan Kettering Cancer Center, we aimed to identify clinical variables associated with COVID-19 severity and assess lymphocyte populations. METHODS. We retrospectively investigated patients diagnosed between March 15th and May 7th, 2020. In a subset of patients, lymphocyte immunophenotyping, quantitative real-time PCR from nasopharyngeal swabs, and SARS-CoV-2 antibody status were available. RESULTS. We identified 77 SARS-CoV-2 + cellular therapy recipients (Allo = 35, Auto = 37, CAR-T = 5; median time from cellular therapy 782 days (IQR 354,1611). Overall survival at 30 days was 78%. Clinical variables significantly associated with the composite endpoint of non-rebreather or higher oxygen requirement and death (n events = 25/77) included number of co-morbidities (HR 5.41, P = 0.004), infiltrates (HR 3.08, P = 0.032), and neutropenia (HR 1.15, P = 0.04). Worsening graft-versus-host-disease was not identified among Allo subjects. Immune profiling revealed reductions and rapid recovery in lymphocyte populations across lymphocyte subsets. Antibody responses were seen in a subset of patients. CONCLUSION. In this series of Allo, Auto, and CAR-T recipients, we report overall favorable clinical outcomes for COVID-19 patients without active malignancy and provide preliminary insights into the lymphocyte populations that are key for the anti-viral response and immune reconstitution. FUNDING. NIH P01 CA23766, NIH/NCI P30 CA008748.

Authors

Gunjan L. Shah, Susan DeWolf, Yeon Joo Lee, Roni Tamari, Parastoo B. Dahi, Jessica A. Lavery, Josel D. Ruiz, Sean M. Devlin, Christina Cho, Jonathan U. Peled, Ioannis Politikos, Michael Scordo, N. Esther Babady, Tania Jain, Santosha Vardhana, Anthony F. Daniyan, Craig S. Sauter, Juliet N. Barker, Sergio A. Giralt, Cheryl Goss, Peter Maslak, Tobias M. Hohl, Mini Kamboj, Lakshmi Ramanathan, Marcel R.M. van den Brink, Esperanza B. Papadopoulos, Genovefa A. Papanicolaou, Miguel-Angel Perales

×

Peripheral CD4+ T cell subsets and antibody response in COVID-19 convalescent individuals
Fang Gong, … , Di Yu, Pengcheng Zhou
Fang Gong, … , Di Yu, Pengcheng Zhou
Published August 25, 2020
Citation Information: J Clin Invest. 2020. https://doi.org/10.1172/JCI141054.
View: Text | PDF

Peripheral CD4+ T cell subsets and antibody response in COVID-19 convalescent individuals

  • Text
  • PDF
Abstract

Background: Marked progress is achieved in understanding the physiopathology of COVID-19 that caused global pandemics. However, CD4+ T cell population that is critical for antibody response in COVID-19 is poorly understood. Methods: In this study, we provided a comprehensive analysis of peripheral CD4+ T cells of 13 COVID-19 convalescent patients, as defined as confirmed free of SARS-CoV-2 for 2-4 weeks, using flow cytometry, magnetic chemiluminescence enzyme antibody immunoassay and correlated the data with clinical characteristics. Results: We observed that relative to healthy individuals, convalescent patients displayed an altered peripheral CD4+ T cell spectrum. Specifically, consistent with other viral infections, cTFH1 cell associated with SARS-CoV-2 targeting antibodies, which was found to skew with disease severity as more severe individuals showed higher frequency of TEM and TFH-EM cells but a lower frequency of TCM, TFH-CM and TNaive cells, relative to mild and moderate patients. Interestingly, higher frequency of cTFH-EM cells correlated with lower number of recorded admission blood oxygen level in convalescent patients. These observations might constitute residual effects by which COVID-19 can impact the homeostasis of CD4+ T cells in the long-term and explain the highest ratio of class-switched virus-specific antibody producing individuals found in our severe COVID-19 cohort. Conclusion: Together, our study demonstrated close connection between CD4+ T cells and antibody production in COVID-19 convalescents.Funding: This study was supported by Six Talent Peaks Project in Jiangsu Province and the National Natural Science Foundation of China (NSFC) grants 81970759.

Authors

Fang Gong, Yaping Dai, Ting Zheng, Liang Cheng, Dan Zhao, Hao Wang, Min Liu, Hao Pei, Tengchuan Jin, Di Yu, Pengcheng Zhou

×

Mapping mania symptoms based on focal brain damage
Gonçalo Cotovio, … , Albino J. Oliveira-Maia, Michael D. Fox
Gonçalo Cotovio, … , Albino J. Oliveira-Maia, Michael D. Fox
Published August 24, 2020
Citation Information: J Clin Invest. 2020. https://doi.org/10.1172/JCI136096.
View: Text | PDF

Mapping mania symptoms based on focal brain damage

  • Text
  • PDF
Abstract

BACKGROUND Although mania is characteristic of bipolar disorder, it can also occur following focal brain damage. Such cases may provide unique insight into brain regions responsible for mania symptoms and identify therapeutic targets.METHODS Lesion locations associated with mania were identified using a systematic literature search (n = 41) and mapped onto a common brain atlas. The network of brain regions functionally connected to each lesion location was computed using normative human connectome data (resting-state functional MRI, n = 1000) and contrasted with those obtained from lesion locations not associated with mania (n = 79). Reproducibility was assessed using independent cohorts of mania lesions derived from clinical chart review (n = 15) and of control lesions (n = 490). Results were compared with brain stimulation sites previously reported to induce or relieve mania symptoms.RESULTS Lesion locations associated with mania were heterogeneous and no single brain region was lesioned in all, or even most, cases. However, these lesion locations showed a unique pattern of functional connectivity to the right orbitofrontal cortex, right inferior temporal gyrus, and right frontal pole. This connectivity profile was reproducible across independent lesion cohorts and aligned with the effects of therapeutic brain stimulation on mania symptoms.CONCLUSIONS Brain lesions associated with mania are characterized by a specific pattern of brain connectivity that lends insight into localization of mania symptoms and potential therapeutic targets.FUNDING Fundação para a Ciência e Tecnologia (FCT), Harvard Medical School DuPont-Warren Fellowship, Portuguese national funds from FCT and Fundo Europeu de Desenvolvimento Regional, Child Neurology Foundation Shields Research, Sidney R. Baer, Jr. Foundation, Nancy Lurie Marks Foundation, Mather’s Foundation, and the NIH.

Authors

Gonçalo Cotovio, Daniel Talmasov, J. Bernardo Barahona-Corrêa, Joey Hsu, Suhan Senova, Ricardo Ribeiro, Louis Soussand, Ana Velosa, Vera Cruz e Silva, Natalia Rost, Ona Wu, Alexander L. Cohen, Albino J. Oliveira-Maia, Michael D. Fox

×

Mass cytometry detects H3.3K27M-specific vaccine responses in diffuse midline glioma
Sabine Mueller, … , Michael D. Prados, Hideho Okada
Sabine Mueller, … , Michael D. Prados, Hideho Okada
Published August 20, 2020
Citation Information: J Clin Invest. 2020. https://doi.org/10.1172/JCI140378.
View: Text | PDF | Corrigendum

Mass cytometry detects H3.3K27M-specific vaccine responses in diffuse midline glioma

  • Text
  • PDF
Abstract

Background: Patients with diffuse midline gliomas (DMG), including diffuse intrinsic pontine glioma (DIPG), have dismal outcomes. We previously described the H3.3K27M mutation as a shared neoantigen in HLA-A*02.01+ H3.3K27M+ DMGs. Within the Pacific Pediatric Neuro-Oncology Consortium, we assessed safety and efficacy of an H3.3K27M-targeted peptide vaccine. Patients and Methods: Newly diagnosed patients aged 3-21 years with HLA-A*02.01+ and H3.3K27M+ status were enrolled into Stratum A (DIPG) and Stratum B (non-pontine DMG). Vaccine was administered in combination with poly-ICLC every three weeks for eight cycles, followed by once every six weeks. Immuno-monitoring and imaging occurred every three months. Imaging was centrally reviewed. Immunological responses were assessed in peripheral blood mononuclear cells using mass cytometry. Results: 19 patients enrolled in Stratum A (median age=11 years) and 10 in Stratum B (median age=13 years). There were no grade 4 treatment-related adverse events (TRAE). Injection site reaction was the most commonly reported TRAE. Overall survival (OS) at 12 months was 40% (95% CI, 22% to 73%) for Stratum A and 39% (95% CI, 16% to 93%) for Stratum B. The median OS was 16.1 months in patients exhibiting an expansion of H3.3K27M-reactive CD8+ T-cells compared to 9.8 months for their counterparts (p=0.05). DIPG patients with below-median baseline levels of myeloid-derived suppressor cells had prolonged OS compared to their counterparts (p<0.01). Immediate pre-treatment dexamethasone administration inversely associated with H3.3K27M-reactive CD8+ T-cell responses. Conclusion: Administration of the H3.3K27M-specific vaccine is well tolerated. Patients with H3.3K27M-specific CD8+ immunological responses demonstrated prolonged OS compared to non-responders.

Authors

Sabine Mueller, Jared M. Taitt, Javier E. Villanueva-Meyer, Erin R. Bonner, Takahide Nejo, Rishi R Lulla, Stewart Goldman, Anu Banerjee, Susan N. Chi, Nicholas S. Whipple, John R. Crawford, Karen Gauvain, Kellie J. Nazemi, Payal B. Watchmaker, Neil D. Almeida, Kaori Okada, Andres M. Salazar, Ryan D. Gilbert, Javad Nazarian, Annette M. Molinaro, Lisa H. Butterfield, Michael D. Prados, Hideho Okada

×
  • ← Previous
  • 1
  • 2
  • …
  • 22
  • 23
  • 24
  • …
  • 38
  • 39
  • Next →

No posts were found with this tag.

Advertisement

Copyright © 2025 American Society for Clinical Investigation
ISSN: 0021-9738 (print), 1558-8238 (online)

Sign up for email alerts