Network


Latest external collaboration on country level. Dive into details by clicking on the dots.

Hotspot


Dive into the research topics where Romain Remark is active.

Publication


Featured researches published by Romain Remark.


Cancer Research | 2014

Dendritic Cells in Tumor-Associated Tertiary Lymphoid Structures Signal a Th1 Cytotoxic Immune Contexture and License the Positive Prognostic Value of Infiltrating CD8+ T Cells

Jeremy Goc; Claire Germain; Thi Kim Duy Vo-Bourgais; Audrey Lupo; Christophe Klein; Samantha Knockaert; Luc de Chaisemartin; Hanane Ouakrim; Etienne Becht; Marco Alifano; Pierre Validire; Romain Remark; Scott A. Hammond; Isabelle Cremer; Diane Damotte; Wolf-Herman Fridman; Marie-Caroline Dieu-Nosjean

Tumor-infiltrating T cells, particularly CD45RO(+)CD8(+) memory T cells, confer a positive prognostic value in human cancers. However, the mechanisms that promote a protective T-cell response in the tumor microenvironment remain unclear. In chronic inflammatory settings such as the tumor microenvironment, lymphoid neogenesis can occur to create local lymph node-like structures known as tertiary lymphoid structures (TLS). These structures can exacerbate a local immune response, such that TLS formation in tumors may help promote an efficacious immune contexture. However, the role of TLS in tumors has yet to be investigated carefully. In lung tumors, mature dendritic cells (DC) present in tumor-associated TLS can provide a specific marker of these structures. In this study, we evaluated the influence of TLS on the characteristics of the immune infiltrate in cohorts of prospective and retrospective human primary lung tumors (n = 458). We found that a high density of mature DC correlated closely to a strong infiltration of T cells that are predominantly of the effector-memory phenotype. Moreover, mature DC density correlated with expression of genes related to T-cell activation, T-helper 1 (Th1) phenotype, and cytotoxic orientation. Lastly, a high density of TLS-associated DC correlated with long-term survival, which also allowed a distinction of patients with high CD8(+) T-cell infiltration but a high risk of death. Taken together, our results show how tumors infiltrated by TLS-associated mature DC generate a specific immune contexture characterized by a strong Th1 and cytotoxic orientation that confers the lowest risk of death. Furthermore, our findings highlight the pivotal function of TLS in shaping the immune character of the tumor microenvironment, in promoting a protective immune response mediated by T cells against cancer.


American Journal of Respiratory and Critical Care Medicine | 2014

Presence of B Cells in Tertiary Lymphoid Structures Is Associated with a Protective Immunity in Patients with Lung Cancer

Claire Germain; Sacha Gnjatic; Fella Tamzalit; Samantha Knockaert; Romain Remark; Jeremy Goc; Alice Lepelley; Etienne Becht; Sandrine Katsahian; Geoffray Bizouard; Pierre Validire; Diane Damotte; Marco Alifano; Pierre Magdeleinat; Isabelle Cremer; Jean-Luc Teillaud; Wolf-Herman Fridman; Marie-Caroline Dieu-Nosjean

RATIONALE It is now well established that immune responses can take place outside of primary and secondary lymphoid organs. We previously described the presence of tertiary lymphoid structures (TLS) in patients with non-small cell lung cancer (NSCLC) characterized by clusters of mature dendritic cells (DCs) and T cells surrounded by B-cell follicles. We demonstrated that the density of these mature DCs was associated with favorable clinical outcome. OBJECTIVES To study the role of follicular B cells in TLS and the potential link with a local humoral immune response in patients with NSCLC. METHODS The cellular composition of TLS was investigated by immunohistochemistry. Characterization of B-cell subsets was performed by flow cytometry. A retrospective study was conducted in two independent cohorts of patients. Antibody specificity was analyzed by ELISA. MEASUREMENTS AND MAIN RESULTS Consistent with TLS organization, all stages of B-cell differentiation were detectable in most tumors. Germinal center somatic hypermutation and class switch recombination machineries were activated, associated with the generation of plasma cells. Approximately half of the patients showed antibody reactivity against up to 7 out of the 33 tumor antigens tested. A high density of follicular B cells correlated with long-term survival, both in patients with early-stage NSCLC and with advanced-stage NSCLC treated with chemotherapy. The combination of follicular B cell and mature DC densities allowed the identification of patients with the best clinical outcome. CONCLUSIONS B-cell density represents a new prognostic biomarker for NSCLC patient survival, and makes the link between TLS and a protective B cell-mediated immunity.


Immunity | 2016

Expansion and Activation of CD103+ Dendritic Cell Progenitors at the Tumor Site Enhances Tumor Responses to Therapeutic PD-L1 and BRAF Inhibition

Hélène Salmon; Juliana Idoyaga; Adeeb Rahman; Marylene Leboeuf; Romain Remark; Stefan Jordan; Maria Casanova-Acebes; Makhzuna Khudoynazarova; Judith Agudo; Navpreet Tung; Svetoslav Chakarov; Christina Rivera; Brandon Hogstad; Marcus Bosenberg; Daigo Hashimoto; Sacha Gnjatic; Nina Bhardwaj; Anna Karolina Palucka; Brian D. Brown; Joshua Brody; Florent Ginhoux; Miriam Merad

Large numbers of melanoma lesions develop resistance to targeted inhibition of mutant BRAF or fail to respond to checkpoint blockade. We explored whether modulation of intratumoral antigen-presenting cells (APCs) could increase responses to these therapies. Using mouse melanoma models, we found that CD103(+) dendritic cells (DCs) were the only APCs transporting intact antigens to the lymph nodes and priming tumor-specific CD8(+) T cells. CD103(+) DCs were required to promote anti-tumoral effects upon blockade of the checkpoint ligand PD-L1; however, PD-L1 inhibition only led to partial responses. Systemic administration of the growth factor FLT3L followed by intratumoral poly I:C injections expanded and activated CD103(+) DC progenitors in the tumor, enhancing responses to BRAF and PD-L1 blockade and protecting mice from tumor rechallenge. Thus, the paucity of activated CD103(+) DCs in tumors limits checkpoint-blockade efficacy and combined FLT3L and poly I:C therapy can enhance tumor responses to checkpoint and BRAF blockade.


American Journal of Respiratory and Critical Care Medicine | 2015

The Non–Small Cell Lung Cancer Immune Contexture. A Major Determinant of Tumor Characteristics and Patient Outcome

Romain Remark; Christian Becker; Jorge E. Gomez; Diane Damotte; Marie-Caroline Dieu-Nosjean; Wolf-Herman Fridman; Charles A. Powell; Nasser K. Altorki; Miriam Merad; Sacha Gnjatic

Solid tumors, beyond mere accumulation of cancer cells, form a complex ecosystem consisting of normal epithelial cells, fibroblasts, blood and lymphatic vessels, structural components, and infiltrating hematopoietic cells including myeloid and lymphoid elements that impact tumor growth, tumor spreading, and clinical outcome. The composition of the immune microenvironment is diverse, including various populations of T cells, B cells, dendritic cells, natural killer cells, myeloid-derived suppressor cells, neutrophils, or macrophages. The immune contexture describes the density, location, and organization of these immune cells within solid tumors. In lung cancer, which is the deadliest type of cancer, and particularly in non-small cell lung cancer, its most prevalent form, reports have described some of the interactions between the tumor and the host. These data, in addition to articles on various types of tumors, provide a greater understanding of the tumor-host microenvironment interaction and stimulate the development of prognostic and predictive biomarkers, the identification of novel target antigens for therapeutic intervention, and the implementation of tools for long-term management of patients with cancer.


Science immunology | 2016

In-depth tissue profiling using multiplexed immunohistochemical consecutive staining on single slide

Romain Remark; Taha Merghoub; Niels Grabe; Geert J. S. Litjens; Diane Damotte; Jedd D. Wolchok; Miriam Merad; Sacha Gnjatic

Multidimensional immunohistochemistry generates high-resolution snapshots of immune cells within a tumor. Putting immune cells on the map Multiplexed flow cytometry allows for careful characterization of the complexity of immune cells. However, these cells do not act in isolation, and multiplexed analyses do not extend to spatial visualization of cells in tissue microenvironments. Remark et al. have combined immunohistochemistry with virtual landscaping to generate maps of immune cells in tumors stained with up to 10 distinct antibodies. Rather than using filters to resolve chromogens, they use multiple rounds of staining and destaining, followed by virtual color assignment. This simple yet powerful technique can be used to chart maps of cells in any tissue. Despite remarkable recent achievements of immunotherapy strategies in cancer treatment, clinical responses remain limited to subsets of patients. Predictive markers of disease course and response to immunotherapy are urgently needed. Recent results have revealed the potential predictive value of immune cell phenotype and spatial distribution at the tumor site, prompting the need for multidimensional immunohistochemical analyses of tumor tissues. To address this need, we developed a sample-sparing, highly multiplexed immunohistochemistry technique based on iterative cycles of tagging, image scanning, and destaining of chromogenic substrate on a single slide. This assay, in combination with a newly developed automated digital landscaping solution, democratizes access to high-dimensional immunohistochemical analyses by capturing the complexity of the immunome using routine pathology standards. Applications of the method extend beyond cancer to screen and validate comprehensive panels of tissue-based prognostic and predictive markers, perform in-depth in situ monitoring of therapies, and identify targets of disease.


Scientific Reports | 2017

Quantification of hepatocellular carcinoma heterogeneity with multiparametric magnetic resonance imaging

Stefanie J. C. G. Hectors; Mathilde Wagner; Octavia Bane; Cecilia Besa; Sara Lewis; Romain Remark; Nelson Chen; M. Isabel Fiel; Hongfa Zhu; Sacha Gnjatic; Miriam Merad; Yujin Hoshida

Tumour heterogeneity poses a significant challenge for treatment stratification. The goals of this study were to quantify heterogeneity in hepatocellular carcinoma (HCC) using multiparametric magnetic resonance imaging (mpMRI), and to report preliminary data correlating quantitative MRI parameters with advanced histopathology and gene expression in a patient subset. Thirty-two HCC patients with 39 HCC lesions underwent mpMRI including diffusion-weighted imaging (DWI), blood-oxygenation-level-dependent (BOLD), tissue-oxygenation-level-dependent (TOLD) and dynamic contrast-enhanced (DCE)-MRI. Histogram characteristics [central tendency (mean, median) and heterogeneity (standard deviation, kurtosis, skewness) MRI parameters] in HCC and liver parenchyma were compared using Wilcoxon signed-rank tests. Histogram data was correlated between MRI methods in all patients and with histopathology and gene expression in 14 patients. HCCs exhibited significantly higher intra-tissue heterogeneity vs. liver with all MRI methods (P < 0.030). Although central tendency parameters showed significant correlations between MRI methods and with each of histopathology and gene expression, heterogeneity parameters exhibited additional complementary correlations between BOLD and DCE-MRI and with histopathologic hypoxia marker HIF1α and gene expression of Wnt target GLUL, pharmacological target FGFR4, stemness markers EPCAM and KRT19 and immune checkpoint PDCD1. Histogram analysis combining central tendency and heterogeneity mpMRI features is promising for non-invasive HCC characterization on the imaging, histologic and genomics levels.


Oncotarget | 2017

MAGE expression in head and neck squamous cell carcinoma primary tumors, lymph node metastases and respective recurrences-implications for immunotherapy

Simon Laban; Gregor Giebel; Niklas Klümper; Andreas Schröck; Johannes Doescher; Giulio C. Spagnoli; Julia Thierauf; Marie-Nicole Theodoraki; Romain Remark; Sacha Gnjatic; Rosemarie Krupar; Andrew G. Sikora; Geert J. S. Litjens; Niels Grabe; Glen Kristiansen; Friedrich Bootz; Patrick J. Schuler; Cornelia Brunner; Johannes Brägelmann; Thomas K. Hoffmann; Sven Perner

Melanoma associated antigens (MAGE) are potential targets for immunotherapy and have been associated with poor overall survival (OS) in head and neck squamous cell carcinoma (HNSCC). However, little is known about MAGE in lymph node metastases (LNM) and recurrent disease (RD) of HNSCC. To assess whether MAGE expression increases with metastasis or recurrence, a tissue microarray (TMA) of 552 primary tumors (PT), 219 LNM and 75 RD was evaluated by immunohistochemistry for MAGE antigens using three monoclonal antibodies to multiple MAGE family members. Mean expression intensity (MEI) was obtained from triplicates of each tumor specimen. The median MEI compared between PT, LNM and RD was significantly higher in LNM and RD. In paired samples, MEI was comparable in PT to respective LNM, but significantly different from RD. Up to 25% of patients were negative for pan-MAGE or MAGE-A3/A4 in PT, but positive in RD. The prognostic impact of MAGE expression was validated in the TMA cohort and also in TCGA data (mRNA). OS was significantly lower for patients expressing pan-MAGE or MAGE-A3/A4 in both independent cohorts. MAGE expression was confirmed as a prognostic marker in HNSCC and may be important for immunotherapeutic strategies as a shared antigen.


Journal of Experimental Medicine | 2018

RAF/MEK/extracellular signal–related kinase pathway suppresses dendritic cell migration and traps dendritic cells in Langerhans cell histiocytosis lesions

Brandon Hogstad; Marie-Luise Berres; Rikhia Chakraborty; Jun Tang; Camille Bigenwald; Madhavika N. Serasinghe; Karen Phaik Har Lim; Howard Lin; Tsz-Kwong Man; Romain Remark; Samantha Baxter; Veronika Kana; Stefan Jordan; Zoi Karoulia; Wing-hong Kwan; Marylene Leboeuf; Elisa Brandt; Hélène Salmon; Kenneth L. McClain; Poulikos I. Poulikakos; Jerry E. Chipuk; Willem J. M. Mulder; Carl E. Allen; Miriam Merad

Langerhans cell histiocytosis (LCH) is an inflammatory myeloid neoplasia characterized by granulomatous lesions containing pathological CD207+ dendritic cells (DCs) with constitutively activated mitogen-activated protein kinase (MAPK) pathway signaling. Approximately 60% of LCH patients harbor somatic BRAFV600E mutations localizing to CD207+ DCs within lesions. However, the mechanisms driving BRAFV600E+ LCH cell accumulation in lesions remain unknown. Here we show that sustained extracellular signal–related kinase activity induced by BRAFV600E inhibits C-C motif chemokine receptor 7 (CCR7)–mediated DC migration, trapping DCs in tissue lesions. Additionally, BRAFV600E increases expression of BCL2-like protein 1 (BCL2L1) in DCs, resulting in resistance to apoptosis. Pharmacological MAPK inhibition restores migration and apoptosis potential in a mouse LCH model, as well as in primary human LCH cells. We also demonstrate that MEK inhibitor-loaded nanoparticles have the capacity to concentrate drug delivery to phagocytic cells, significantly reducing off-target toxicity. Collectively, our results indicate that MAPK tightly suppresses DC migration and augments DC survival, rendering DCs in LCH lesions trapped and resistant to cell death.


Cancer immunology research | 2016

Abstract B109: In-depth tissue analysis using multiplexed immunohistochemical consecutive staining on single slide

Romain Remark; Taha Merghoub; Diane Damotte; Jedd D. Wolchok; Miriam Merad; Sacha Gnjatic

Recent successes of cancer immunotherapy, in particular immune checkpoint blockade, have been largely attributed to factors in the tumor microenvironment (TME). In addition, many studies have found that high lymphocyte infiltration in tumors is prognostic of progression-free or overall survival, while in some cases, myeloid cells rather than lymphocytes are associated with favorable clinical outcome. Importantly, assessment of immune markers usually requires not only quantifying their presence in tumors but also assessing their spatial localization. To assess spatial distribution of tumor-infiltrating immune cells and suppressive molecules, multidimensional immunohistochemical analyses are poised to become the new standard in pathology. A major limitation for such high-dimensional analyses however is tumor tissue availability and new multiplexed imaging approaches are desperately needed. Here, we will describe the development of a new multiplexed immunohistochemistry (IHC) method independent of proprietary reagents or equipment to assess a large panel of phenotypical markers that could easily be integrated in routine clinical pathology. This new technique named Multiplexed Immunohistochemical Consecutive Staining on Single Slide (MICSSS) can be performed in any laboratory already conducting traditional IHC with limited cost and no additional instrumentation. The MICSSS method is performed on formalin-fixed paraffin-embedded tissue using iterative cycles of staining, revelation, scanning, and bleaching of chromogenic substrate. It should easily be adaptable to most existing staining protocols with primary antibody staining and secondary antibody detection, thus retaining previously established specificity and sensitivity parameters. The MICSSS method can characterize a large panel of parameters, including marker colocalization on cells, using a single slide while preserving tissue antigenicity and architecture. It is not limited by antibody cross-reactivity, photo-bleaching, or autofluorescence, while still allowing prolonged slide storage for future reuse as new markers become available. Here, we will describe the MICSSS method workflow and show potential applications of multiplex immunostaining including its use to characterize the tumor immune environment and identify prognostic factors or predictive biomarkers of response to CTLA-4 blockade. In conclusion, the MICSSS method will provide new powerful ways to map the tumor microenvironment with precision, in a sample-sparing manner, to monitor the immune response at the tumor site during therapy and discover new prognostic and predictive markers that might be of major interest for the clinical management of patients with various diseases. Citation Format: Romain Remark, Taha Merghoub, Diane Damotte, Jedd D. Wolchok, Miriam Merad, Sacha Gnjatic. In-depth tissue analysis using multiplexed immunohistochemical consecutive staining on single slide. [abstract]. In: Proceedings of the CRI-CIMT-EATI-AACR Inaugural International Cancer Immunotherapy Conference: Translating Science into Survival; September 16-19, 2015; New York, NY. Philadelphia (PA): AACR; Cancer Immunol Res 2016;4(1 Suppl):Abstract nr B109.


Cancer Research | 2017

Abstract 334: PTEN-L regulates epithelial growth and macrophage function

Andrew L. Wolfe; Benjamin Hopkins; Sebastián A. Riquelme; Kipyegon Kitur; Sait Ozturk; Kyeongah Kang; Romain Remark; Adeeb Rahman; Chyuan-Sheng Lin; Miriam Merad; Matthias Szabolcs; Shu-Hsia Chen; Alice Prince; Ramon Parsons

PTEN is among the most frequently mutated and deleted tumor suppressor genes in many malignancies, including breast cancer. An alternatively translated long form of PTEN, termed PTEN-L, has divergent functionality from PTEN, although its function at the organism level has not been studied. Here, we report a knockout mouse with specific ablation of PTEN-L expression but intact expression of PTEN. These mice display mammary ductal hyperplasia characterized by increased luminal growth and increased numbers of macrophages in the surrounding stroma. Macrophages are particularly affected by PTEN-L loss, with significant changes to their secretomes and functional deficiencies in clearing bacterial infections, consistent with a shift toward an M2-like polarization. Overall, these findings demonstrate that PTEN-L has unique functions in regulating mammary epithelial growth and macrophage functionality that are independent of canonical PTEN. Citation Format: Andrew L. Wolfe, Benjamin D. Hopkins, Sebastian A. Riquelme, Kipyegon Kitur, Sait Ozturk, Kyeongah Kang, Romain Remark, Adeeb Rahman, Chyuan-Sheng Lin, Miriam Merad, Matthias Szabolcs, Shu-Hsia Chen, Alice Prince, Ramon Parsons. PTEN-L regulates epithelial growth and macrophage function [abstract]. In: Proceedings of the American Association for Cancer Research Annual Meeting 2017; 2017 Apr 1-5; Washington, DC. Philadelphia (PA): AACR; Cancer Res 2017;77(13 Suppl):Abstract nr 334. doi:10.1158/1538-7445.AM2017-334

Collaboration


Dive into the Romain Remark's collaboration.

Top Co-Authors

Avatar

Sacha Gnjatic

Icahn School of Medicine at Mount Sinai

View shared research outputs
Top Co-Authors

Avatar

Miriam Merad

Icahn School of Medicine at Mount Sinai

View shared research outputs
Top Co-Authors

Avatar

Adeeb Rahman

Icahn School of Medicine at Mount Sinai

View shared research outputs
Top Co-Authors

Avatar

Diane Damotte

Paris Descartes University

View shared research outputs
Top Co-Authors

Avatar

Camille Bigenwald

Icahn School of Medicine at Mount Sinai

View shared research outputs
Top Co-Authors

Avatar

Christian Becker

Icahn School of Medicine at Mount Sinai

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

El-ad David Amir

Icahn School of Medicine at Mount Sinai

View shared research outputs
Top Co-Authors

Avatar

Hélène Salmon

Icahn School of Medicine at Mount Sinai

View shared research outputs
Top Co-Authors

Avatar

Marco Alifano

Paris Descartes University

View shared research outputs
Researchain Logo
Decentralizing Knowledge