Matthias Gromeier

Overview:

I am a classically trained virologist with a focus on molecular mechanisms of RNA virus pathogenesis. My career is dedicated to unraveling RNA virus:host relations and devising methods of exploiting them for cancer immunotherapy and vaccine design. My background is in translation regulation and mRNA metabolism, viral RNA sensing and innate immunity, and cancer immunology and immunotherapy. Basic mechanistic research in my laboratory is supporting an ambitious clinical translational research program with active multi-center clinical trials in several cancer indications. 

Positions:

Professor of Neurosurgery

Neurosurgery, Neuro-Oncology
School of Medicine

Professor in Molecular Genetics and Microbiology

Molecular Genetics and Microbiology
School of Medicine

Professor in Medicine

Medicine, Infectious Diseases
School of Medicine

Professor of Cell Biology

Cell Biology
School of Medicine

Member of the Duke Cancer Institute

Duke Cancer Institute
School of Medicine

Education:

M.D. 1992

University of Hamburg (Germany)

Postdoctoral fellow, Molecular Genetics & Microbiology

State University of New York, Stony Brook

Postdoctoral Associate, Molecular Genetics & Microbiology

State University of New York, Stony Brook

Grants:

Recombinant Attenuated Poliovirus Immunization Vectors Targeting H3.3(K27M) in DIPG

Administered By
Neurosurgery, Neuro-Oncology Clinical Research
Awarded By
Michael Mosier Defeat DIPG Foundation
Role
Co Investigator
Start Date
End Date

Oncolytic PVSRIPO Expressing Tumor Antigens as a Cancer Vaccine

Administered By
Neurosurgery
Awarded By
Oligo Nation
Role
Principal Investigator
Start Date
End Date

Oncolytic Poliovirus Immunotherapy of Malignant Glioma

Administered By
Pathology
Awarded By
The Slomo and Cindy Silvian Foundation, Inc
Role
Co Investigator
Start Date
End Date

Oncolytic Virotherapy of Meningeal Cancer

Administered By
Neurosurgery, Neuro-Oncology
Awarded By
National Institutes of Health
Role
Principal Investigator
Start Date
End Date

Establishing a Rationale for PVSRIPO Immunotherapy in Newly Diagnosed GBM

Administered By
Neurosurgery
Awarded By
National Brain Tumor Society
Role
Principal Investigator
Start Date
End Date

Publications:

Intratumor childhood vaccine-specific CD4+ T-cell recall coordinates antitumor CD8+ T cells and eosinophils.

BACKGROUND: Antitumor mechanisms of CD4+ T cells remain crudely defined, and means to effectively harness CD4+ T-cell help for cancer immunotherapy are lacking. Pre-existing memory CD4+ T cells hold potential to be leveraged for this purpose. Moreover, the role of pre-existing immunity in virotherapy, particularly recombinant poliovirus immunotherapy where childhood polio vaccine specific immunity is ubiquitous, remains unclear. Here we tested the hypothesis that childhood vaccine-specific memory T cells mediate antitumor immunotherapy and contribute to the antitumor efficacy of polio virotherapy. METHODS: The impact of polio immunization on polio virotherapy, and the antitumor effects of polio and tetanus recall were tested in syngeneic murine melanoma and breast cancer models. CD8+ T-cell and B-cell knockout, CD4+ T-cell depletion, CD4+ T-cell adoptive transfer, CD40L blockade, assessments of antitumor T-cell immunity, and eosinophil depletion defined antitumor mechanisms of recall antigens. Pan-cancer transcriptome data sets and polio virotherapy clinical trial correlates were used to assess the relevance of these findings in humans. RESULTS: Prior vaccination against poliovirus substantially bolstered the antitumor efficacy of polio virotherapy in mice, and intratumor recall of poliovirus or tetanus immunity delayed tumor growth. Intratumor recall antigens augmented antitumor T-cell function, caused marked tumor infiltration of type 2 innate lymphoid cells and eosinophils, and decreased proportions of regulatory T cells (Tregs). Antitumor effects of recall antigens were mediated by CD4+ T cells, limited by B cells, independent of CD40L, and dependent on eosinophils and CD8+ T cells. An inverse relationship between eosinophil and Treg signatures was observed across The Cancer Genome Atlas (TCGA) cancer types, and eosinophil depletion prevented Treg reductions after polio recall. Pretreatment polio neutralizing antibody titers were higher in patients living longer, and eosinophil levels increased in the majority of patients, after polio virotherapy. CONCLUSION: Pre-existing anti-polio immunity contributes to the antitumor efficacy of polio virotherapy. This work defines cancer immunotherapy potential of childhood vaccines, reveals their utility to engage CD4+ T-cell help for antitumor CD8+ T cells, and implicates eosinophils as antitumor effectors of CD4+ T cells.
MLA Citation
Brown, Michael C., et al. “Intratumor childhood vaccine-specific CD4+ T-cell recall coordinates antitumor CD8+ T cells and eosinophils.J Immunother Cancer, vol. 11, no. 4, Apr. 2023. Pubmed, doi:10.1136/jitc-2022-006463.
URI
https://scholars.duke.edu/individual/pub1573124
PMID
37072349
Source
pubmed
Published In
Journal for Immunotherapy of Cancer
Volume
11
Published Date
DOI
10.1136/jitc-2022-006463

CD155 is a putative therapeutic target in medulloblastoma.

BACKGROUND: Medulloblastoma is the most common pediatric malignant brain tumor, consisting of four molecular subgroups (WNT, SHH, Group 3, Group 4) and 12 subtypes. Expression of the cell surface poliovirus receptor (PVR), CD155, is necessary for entry of the viral immunotherapeutic agent, PVSRIPO, a polio:rhinovirus chimera. CD155, physiologically expressed in the mononuclear phagocytic system, is widely expressed ectopically in solid tumors. The objective of this study is to elucidate CD155 expression as both a receptor for PVSRIPO and a therapeutic target in medulloblastoma. METHODS: PVR mRNA expression was determined in several patient cohorts and human medulloblastoma cell lines. Patient samples were also analyzed for CD155 expression using immunohistochemistry and cell lines were analyzed using Western Blots. CD155 was blocked using a monoclonal antibody and cell viability, invasion, and migration were assessed. RESULTS AND DISCUSSION: PVR mRNA expression was highest in the WNT subgroup and lowest in Group 4. PVR expression in the subgroups of medulloblastoma were similar to other pediatric brain and non-brain tumors. PVR expression was largely not associated with subgroup or subtype. Neither PVR protein expression intensity nor frequency were associated with overall survival. PVR expression was elevated in Group 3 patients with metastases but there was no difference in paired primary and metastatic medulloblastoma. Blocking PVR resulted in dose-dependent cell death, decreased invasion in vitro, and modestly inhibited cell migration. CONCLUSIONS: CD155 is expressed across medulloblastoma subgroups and subtypes. Blocking CD155 results in cell death and decreased cellular invasion. This study provides rationale for CD155-targeting agents including PVSRIPO and antibody-mediated blockade of CD155.
Authors
Li, S; McLendon, R; Sankey, E; Kornahrens, R; Lyne, A-M; Cavalli, FMG; McKay, Z; Herndon, JE; Remke, M; Picard, D; Gromeier, M; Brown, M; Thompson, EM
MLA Citation
Li, Sean, et al. “CD155 is a putative therapeutic target in medulloblastoma.Clin Transl Oncol, vol. 25, no. 3, Mar. 2023, pp. 696–705. Pubmed, doi:10.1007/s12094-022-02975-9.
URI
https://scholars.duke.edu/individual/pub1555228
PMID
36301489
Source
pubmed
Published In
Clin Transl Oncol
Volume
25
Published Date
Start Page
696
End Page
705
DOI
10.1007/s12094-022-02975-9

Multimodality analysis confers a prognostic benefit of a T-cell infiltrated tumor microenvironment and peripheral immune status in patients with melanoma.

BACKGROUND: We previously reported results from a phase 1 study testing intratumoral recombinant poliovirus, lerapolturev, in 12 melanoma patients. All 12 patients received anti-PD-1 systemic therapy before lerapolturev, and 11 of these 12 patients also received anti-PD-1 after lerapolturev. In preclinical models lerapolturev induces intratumoral innate inflammation that engages antitumor T cells. In the current study, prelerapolturev and postlerapolturev tumor biopsies and blood were evaluated for biomarkers of response. METHODS: The following analyses were performed on tumor tissue (n=11): (1) flow cytometric assessment of immune cell density, (2) NanoString Digital Spatial profiling of protein and the transcriptome, and (3) bulk RNA sequencing. Immune cell phenotypes and responsiveness to in vitro stimulation, including in vitro lerapolturev challenge, were measured in peripheral blood (n=12). RESULTS: Three patients who received anti-PD-1 therapy within 30 days of lerapolturev have a current median progression-free survival (PFS) of 2.3 years and had higher CD8+T cell infiltrates in prelerapolturev tumor biopsies relative to that of 7 patients with median PFS of 1.6 months and lower CD8+T cell infiltrates in prelerapolturev tumor biopsies. In peripheral blood, four patients with PFS 2.3 years (including three that received anti-PD-1 therapy within 30 days before lerapolturev and had higher pretreatment tumor CD8+T cell infiltrates) had significantly higher effector memory (CD8+, CCR7-, CD45RA-) but lower CD8+PD-1+ and CD4+PD-1+ cells compared with eight patients with median PFS 1.6 months. In addition, pretreatment blood from the four patients with median PFS 2.3 years had more potent antiviral responses to in vitro lerapolturev challenge compared with eight patients with median PFS 1.6 months. CONCLUSION: An inflamed pretreatment tumor microenvironment, possibly induced by prior anti-PD-1 therapy and a proficient peripheral blood pretreatment innate immune response (antiviral/interferon signaling) to lerapolturev was associated with long term PFS after intratumoral lerapolturev in a small cohort of patients. These findings imply a link between intratumoral T cell inflammation and peripheral immune function. TRIAL REGISTRATION NUMBER: NCT03712358.
Authors
Beasley, GM; Brown, MC; Farrow, NE; Landa, K; Al-Rohil, RN; Selim, MA; Therien, AD; Jung, S-H; Gao, J; Boczkowski, D; Holl, EK; Salama, AKS; Bigner, DD; Gromeier, M; Nair, SK
MLA Citation
Beasley, Georgia M., et al. “Multimodality analysis confers a prognostic benefit of a T-cell infiltrated tumor microenvironment and peripheral immune status in patients with melanoma.J Immunother Cancer, vol. 10, no. 9, Sept. 2022. Pubmed, doi:10.1136/jitc-2022-005052.
URI
https://scholars.duke.edu/individual/pub1552997
PMID
36175036
Source
pubmed
Published In
Journal for Immunotherapy of Cancer
Volume
10
Published Date
DOI
10.1136/jitc-2022-005052

STING IS SILENCED IN GBM BY PROMOTER METHYLATION THAT IS ESTABLISHED BY TISSUE OF ORIGIN

Authors
Low, J; Chandramohan, V; Bowie, M; Brown, M; Waitkus, M; Briley, A; Stevenson, K; Fuller, R; Reitman, Z; Muscat, A; Hariharan, S; Hostettler, J; Wong, N; Gromeier, M; Ashley, DM
MLA Citation
Low, Justin, et al. “STING IS SILENCED IN GBM BY PROMOTER METHYLATION THAT IS ESTABLISHED BY TISSUE OF ORIGIN.” Neuro Oncology, vol. 23, 2021, pp. 4–4.
URI
https://scholars.duke.edu/individual/pub1511792
Source
wos-lite
Published In
Neuro Oncology
Volume
23
Published Date
Start Page
4
End Page
4

SAFETY AND EFFICACY OF PVSRIPO IN RECURRENT GLIOBLASTOMA: LONG-TERM FOLLOW-UP AND INITIAL MULTICENTER RESULTS

Authors
Desjardins, A; Gromeier, M; Friedman, H; Landi, D; Friedman, A; Ashley, DM; Curry, W; Butowski, N; Sloan, A; Wen, P; Cavaliere, R; Gerstner, E; Berger, M; Buerki, R; Chiocca, EA; Sauvageau, E; Kelly, A; Nichols, WG; Mixson, L; Dickinson, A; Orr, K; Learn, C; Bigner, D
MLA Citation
Desjardins, Annick, et al. “SAFETY AND EFFICACY OF PVSRIPO IN RECURRENT GLIOBLASTOMA: LONG-TERM FOLLOW-UP AND INITIAL MULTICENTER RESULTS.” Neuro Oncology, vol. 23, 2021, pp. 97–97.
URI
https://scholars.duke.edu/individual/pub1511869
Source
wos-lite
Published In
Neuro Oncology
Volume
23
Published Date
Start Page
97
End Page
97

Research Areas:

Brain--Tumors
Cancer--Immunotherapy
Immunity, Innate
Peptide Chain Initiation, Translational
Vaccines
Virology