Try a new search

Format these results:

Searched for:

person:demars01

in-biosketch:yes

Total Results:

216


CD73 blockade promotes dendritic cell infiltration of irradiated tumors and tumor rejection

Wennerberg, Erik; Spada, Sheila; Rudqvist, Nils-Petter; Lhuillier, Claire; Gruber, Sylvia; Chen, Qiuying; Zhang, Fengli; Zhou, Xi K; Gross, Steven S; Formenti, Silvia C; Demaria, Sandra
The ability of focal radiotherapy to promote priming of tumor-specific CD8+ T cells and increase responses to immunotherapy is dependent on infiltration of the tumor by Batf3-dependent conventional dendritic cell type 1 (cDC1) cells. Such infiltration is driven by radiotherapy-induced IFN type I (IFN-I). Other signals may also modulate cDC1 infiltration of irradiated tumors. Here we found increased expression of adenosine-generating enzymes CD38 and CD73 in irradiated mouse and human breast cancer cells and increased adenosine in mouse tumors following radiotherapy. CD73 blockade alone had no effect. CD73 blockade with radiotherapy restored radiotherapy-induced cDC1 infiltration of tumors in settings where radiotherapy induction of IFN-I was suboptimal. In the absence of radiotherapy-induced IFN-I, blockade of CD73 was required for rejection of the irradiated tumor and for systemic tumor control (abscopal effect) in the context of CTLA-4-blockade. These results suggest that CD73 may be a radiation-induced checkpoint, and that CD73 blockade in combination with radiotherapy and immune checkpoint blockade might improve patient response to therapy.
PMID: 32047024
ISSN: 2326-6074
CID: 4311552

The path to a better biomarker: application of a risk management framework for the implementation of PD-L1 and TILs as immuno-oncology biomarkers into breast cancer clinical trials and daily practice

Gonzalez-Ericsson, Paula I; Stovgaard, Elisabeth S; Sua, Luz F; Reisenbichler, Emily; Kos, Zuzana; Carter, Jodi M; Michiels, Stefan; Le Quesne, John; Nielsen, Torsten O; Laenkholm, Anne-Vibeke; Fox, Stephen B; Adam, Julien; Bartlett, John Ms; Rimm, David L; Quinn, Cecily; Peeters, Dieter; Dieci, Maria V; Vincent-Salomon, Anne; Cree, Ian; Hida, Akira I; Balko, Justin M; Haynes, Harry R; Frahm, Isabel; Acosta-Haab, Gabriela; Balancin, Marcelo; Bellolio, Enrique; Yang, Wentao; Kirtani, Pawan; Sugie, Tomoharu; Ehinger, Anna; Castaneda, Carlos A; Kok, Marleen; McArthur, Heather; Siziopikou, Kalliopi; Badve, Sunil; Fineberg, Susan; Gown, Allen; Viale, Giuseppe; Schnitt, Stuart J; Pruneri, Giancarlo; Penault-Llorca, Frederique; Hewitt, Stephen; Thompson, E Aubrey; Allison, Kimberly H; Symmans, William F; Bellizzi, Andrew M; Brogi, Edi; Moore, David A; Larsimont, Denis; Dillon, Deborah A; Lazar, Alexander; Lien, Huangchun; Goetz, Matthew P; Broeckx, Glenn; El Bairi, Khalid; Harbeck, Nadia; Cimino-Mathews, Ashley; Sotiriou, Christos; Adams, Sylvia; Liu, Shi-Wei; Loibl, Sibylle; Chen, I-Chun; Lakhani, Sunil R; Juco, Jonathan W; Denkert, Carsten; Blackley, Elizabeth F; Demaria, Sandra; Leon-Ferre, Roberto; Gluz, Oleg; Zardavas, Dimitrios; Emancipator, Kenneth; Ely, Scott; Loi, Sherene; Salgado, Roberto; Sanders, Melinda
Immune checkpoint inhibitor therapies targeting PD-1/PD-L1 are now standard of care in oncology across several hematologic and solid tumor types, including triple negative breast cancer (TNBC). Patients with metastatic or locally advanced TNBC with PD-L1 expression on immune cells occupying ≥1% of tumor area demonstrated survival benefit with the addition of atezolizumab to nab-paclitaxel. However, concerns regarding variability between immunohistochemical PD-L1 assay performance and inter-reader reproducibility have been raised. High tumor-infiltrating lymphocytes (TILs) have also been associated with response to PD-1/PD-L1 inhibitors in patients with breast cancer. TILs can be easily assessed on hematoxylin and eosin stained slides and have shown reliable inter-reader reproducibility. As an established prognostic factor in early stage TNBC, TILs are soon anticipated to be reported in daily practice in many pathology laboratories worldwide. Since TILs and PD-L1 are parts of an immunological spectrum in breast cancer, we propose the systematic implementation of combined PD-L1 and TIL analyses as a more comprehensive immune-oncological biomarker for patient selection for PD-1/PD-L1 inhibition-based therapy in patients with breast cancer. Although practical and regulatory considerations differ by jurisdiction, the pathology community has the responsibility to patients to implement assays that lead to optimal patient selection. We propose herewith a risk-management framework that may help mitigate the risks of suboptimal patient selection for immuno-therapeutic approaches in clinical trials and daily practice based on combined TILs/PD-L1 assessment in breast cancer. This article is protected by copyright. All rights reserved.
PMID: 32129476
ISSN: 1096-9896
CID: 4339702

Consensus guidelines for the definition, detection and interpretation of immunogenic cell death

Galluzzi, Lorenzo; Vitale, Ilio; Warren, Sarah; Adjemian, Sandy; Agostinis, Patrizia; Martinez, Aitziber Buqué; Chan, Timothy A; Coukos, George; Demaria, Sandra; Deutsch, Eric; Draganov, Dobrin; Edelson, Richard L; Formenti, Silvia C; Fucikova, Jitka; Gabriele, Lucia; Gaipl, Udo S; Gameiro, Sofia R; Garg, Abhishek D; Golden, Encouse; Han, Jian; Harrington, Kevin J; Hemminki, Akseli; Hodge, James W; Hossain, Dewan Md Sakib; Illidge, Tim; Karin, Michael; Kaufman, Howard L; Kepp, Oliver; Kroemer, Guido; Lasarte, Juan Jose; Loi, Sherene; Lotze, Michael T; Manic, Gwenola; Merghoub, Taha; Melcher, Alan A; Mossman, Karen L; Prosper, Felipe; Rekdal, Øystein; Rescigno, Maria; Riganti, Chiara; Sistigu, Antonella; Smyth, Mark J; Spisek, Radek; Stagg, John; Strauss, Bryan E; Tang, Daolin; Tatsuno, Kazuki; van Gool, Stefaan W; Vandenabeele, Peter; Yamazaki, Takahiro; Zamarin, Dmitriy; Zitvogel, Laurence; Cesano, Alessandra; Marincola, Francesco M
Cells succumbing to stress via regulated cell death (RCD) can initiate an adaptive immune response associated with immunological memory, provided they display sufficient antigenicity and adjuvanticity. Moreover, multiple intracellular and microenvironmental features determine the propensity of RCD to drive adaptive immunity. Here, we provide an updated operational definition of immunogenic cell death (ICD), discuss the key factors that dictate the ability of dying cells to drive an adaptive immune response, summarize experimental assays that are currently available for the assessment of ICD in vitro and in vivo, and formulate guidelines for their interpretation.
PMCID:7064135
PMID: 32209603
ISSN: 2051-1426
CID: 4357872

The abscopal effect 67 years later: from a side story to center stage

Demaria, Sandra; Formenti, Silvia C
For over a century, ionising radiation has been used to treat cancer based on its cytotoxic effects on tumour cells. Technical progress has enabled more precise targeting of the tumour to reduce normal tissue toxicity while delivering higher radiation doses per fraction of treatment.In 1953, unexpected regression in lesions outside of the irradiated field were noted by an observant physician, RH Mole, who named such phenomenon "abscopal effect" from the Latin ab (position away from) and scopus (mark or target), in an article published in this journal. Clinical abscopal responses have been reported over the years but because of their very rare occurrence they could not be methodically studied, remaining akin to a curiosity. Nevertheless, their occurrence has ignited interest in studying the systemic effects of radiotherapy. Progress in dissecting the mechanisms that govern the function of the immune system in cancer has enabled to study the implication of immunity in the abscopal effect of radiation. It has become clear that ionising radiation activates canonical pathways of response to viral infections, and can stimulate antitumour immunity. These immune stimulatory effects of radiation have become clinically relevant in the current era of cancer immunotherapy, rendering abscopal responses in patients an attainable aim. Here, we will briefly review the parallel evolutions of two separate fields of medicine, radiation therapy and cancer immunology, and discuss their therapeutic partnership.
PMID: 32101479
ISSN: 1748-880x
CID: 4323452

Aggressive Mammary Cancers Lacking Lymphocytic Infiltration Arise in Irradiated Mice and Can be Prevented by Dietary Intervention

Omene, Coral; Ma, Lin; Moore, Jade; Ouyang, Haoxu; Illa-Bochaca, Irineu; Chou, William; Patel, Manan S; Sebastiano, Christopher; Demaria, Sandra; Mao, Jian-Hua; Karagoz, Kubra; Gatza, Michael L; Barcellos-Hoff, Mary Helen
Because the incidence of breast cancer increases decades after ionizing radiation exposure, aging has been implicated in the evolution of the tumor microenvironment and tumor progression. Here, we investigated radiation-induced carcinogenesis using a model in which the mammary glands of 10-month-old BALB/c mice were transplanted with Trp53-null mammary tissue three days after exposure to low doses of sparsely ionizing γ-radiation or densely ionizing particle radiation. Mammary transplants in aged irradiated hosts gave rise to significantly more tumors that grew more rapidly than those in sham-irradiated mice, with the most pronounced effects seen in mice irradiated with densely ionizing particle radiation. Tumor transcriptomes identified a characteristic immune signature of these aggressive cancers. Consistent with this, fast-growing tumors exhibited an immunosuppressive tumor microenvironment with few infiltrating lymphocytes, abundant immunosuppressive myeloid cells, and high cyclooxgenase-2 and TGFβ. Only aged irradiated hosts gave rise to tumors lacking cytotoxic CD8+ lymphocytes (defined here as immune desert), which also occurred in younger mice. These data suggested that host irradiation may promote immunosuppression. To test this, young chimera mice were fed chow containing a honeybee-derived compound with anti-inflammatory and immunomodulatory properties, caffeic acid phenethyl ester (CAPE). CAPE prevented the detrimental effects of host irradiation on tumor growth rate, immune signature, and immunosuppression. These data indicated that low-dose radiation, particularly densely ionizing radiation, promoted more aggressive cancers by suppressing antitumor immunity. Dietary intervention with a non-toxic immunomodulatory agent could prevent systemic effects of radiation that fuel carcinogenesis, supporting the potential of this strategy for cancer prevention.
PMID: 31831632
ISSN: 2326-6074
CID: 4234912

Exercise reduces immune suppression and breast cancer progression in a preclinical model

Wennerberg, Erik; Lhuillier, Claire; Rybstein, Marissa D; Dannenberg, Kyle; Rudqvist, Nils-Petter; Koelwyn, Graeme J; Jones, Lee W; Demaria, Sandra
Exercise is associated with favorable changes in circulating immune cells and improved survival in early-stage breast cancer patients, but the mechansims remain to be fully elucidated. Preclinical studies indicate that physical activity started before tumor injection reduces tumor incidence and progression. Here we tested whether exercise has anti-tumor effects in mice with established 4T1 mammary carcinoma, a mouse model of triple negative breast cancer. Exercise slowed tumor progression and reduced the tumor-induced accumulation of myeloid-derived suppressor cells (MDSCs). The reduction in MDSCs was accompanied by a relative increase in natural killer and CD8 T cell activation, suggesting that exercise restores a favorable immune environment. Consistently, exercise improved responses to a combination of programmed cell death protein 1 (PD-1) blockade and focal radiotherapy. These data support further investigations of exercise in breast cancer patients treated with combinations of immunotherapy and cytotoxic agents to improve cancer outcomes.
PMCID:6996907
PMID: 32064049
ISSN: 1949-2553
CID: 4311972

Immunogenic Cell Death Driven by Radiation-Impact on the Tumor Microenvironment

Yamazaki, Takahiro; Vanpouille-Box, Claire; Demaria, Sandra; Galluzzi, Lorenzo
Immunogenic cell death (ICD) is a particular form of cell death that can initiate adaptive immunity against antigens expressed by dying cells in the absence of exogenous adjuvants. This implies that cells undergoing ICD not only express antigens that are not covered by thymic tolerance, but also deliver adjuvant-like signals that enable the recruitment and maturation of antigen-presenting cells toward an immunostimulatory phenotype, culminating with robust cross-priming of antigen-specific CD8+ T cells. Such damage-associated molecular patterns (DAMPs), which encompass cellular proteins, small metabolites and cytokines, are emitted in a spatiotemporally defined manner in the context of failing adaptation to stress. Radiation therapy (RT) is a bona fide inducer of ICD, at least when employed according to specific doses and fractionation schedules. Here, we discuss the mechanisms whereby DAMPs emitted by cancer cells undergoing RT-driven ICD alter the functional configuration of the tumor microenvironment.
PMID: 32215874
ISSN: 0927-3042
CID: 4358092

Report on computational assessment of Tumor Infiltrating Lymphocytes from the International Immuno-Oncology Biomarker Working Group

Amgad, Mohamed; Stovgaard, Elisabeth Specht; Balslev, Eva; Thagaard, Jeppe; Chen, Weijie; Dudgeon, Sarah; Sharma, Ashish; Kerner, Jennifer K; Denkert, Carsten; Yuan, Yinyin; AbdulJabbar, Khalid; Wienert, Stephan; Savas, Peter; Voorwerk, Leonie; Beck, Andrew H; Madabhushi, Anant; Hartman, Johan; Sebastian, Manu M; Horlings, Hugo M; Hudeček, Jan; Ciompi, Francesco; Moore, David A; Singh, Rajendra; Roblin, Elvire; Balancin, Marcelo Luiz; Mathieu, Marie-Christine; Lennerz, Jochen K; Kirtani, Pawan; Chen, I-Chun; Braybrooke, Jeremy P; Pruneri, Giancarlo; Demaria, Sandra; Adams, Sylvia; Schnitt, Stuart J; Lakhani, Sunil R; Rojo, Federico; Comerma, Laura; Badve, Sunil S; Khojasteh, Mehrnoush; Symmans, W Fraser; Sotiriou, Christos; Gonzalez-Ericsson, Paula; Pogue-Geile, Katherine L; Kim, Rim S; Rimm, David L; Viale, Giuseppe; Hewitt, Stephen M; Bartlett, John M S; Penault-Llorca, Frédérique; Goel, Shom; Lien, Huang-Chun; Loibl, Sibylle; Kos, Zuzana; Loi, Sherene; Hanna, Matthew G; Michiels, Stefan; Kok, Marleen; Nielsen, Torsten O; Lazar, Alexander J; Bago-Horvath, Zsuzsanna; Kooreman, Loes F S; van der Laak, Jeroen A W M; Saltz, Joel; Gallas, Brandon D; Kurkure, Uday; Barnes, Michael; Salgado, Roberto; Cooper, Lee A D
Assessment of tumor-infiltrating lymphocytes (TILs) is increasingly recognized as an integral part of the prognostic workflow in triple-negative (TNBC) and HER2-positive breast cancer, as well as many other solid tumors. This recognition has come about thanks to standardized visual reporting guidelines, which helped to reduce inter-reader variability. Now, there are ripe opportunities to employ computational methods that extract spatio-morphologic predictive features, enabling computer-aided diagnostics. We detail the benefits of computational TILs assessment, the readiness of TILs scoring for computational assessment, and outline considerations for overcoming key barriers to clinical translation in this arena. Specifically, we discuss: 1. ensuring computational workflows closely capture visual guidelines and standards; 2. challenges and thoughts standards for assessment of algorithms including training, preanalytical, analytical, and clinical validation; 3. perspectives on how to realize the potential of machine learning models and to overcome the perceptual and practical limits of visual scoring.
PMCID:7217824
PMID: 32411818
ISSN: 2374-4677
CID: 4431692

Pitfalls in assessing stromal tumor infiltrating lymphocytes (sTILs) in breast cancer

Kos, Zuzana; Roblin, Elvire; Kim, Rim S; Michiels, Stefan; Gallas, Brandon D; Chen, Weijie; van de Vijver, Koen K; Goel, Shom; Adams, Sylvia; Demaria, Sandra; Viale, Giuseppe; Nielsen, Torsten O; Badve, Sunil S; Symmans, W Fraser; Sotiriou, Christos; Rimm, David L; Hewitt, Stephen; Denkert, Carsten; Loibl, Sibylle; Luen, Stephen J; Bartlett, John M S; Savas, Peter; Pruneri, Giancarlo; Dillon, Deborah A; Cheang, Maggie Chon U; Tutt, Andrew; Hall, Jacqueline A; Kok, Marleen; Horlings, Hugo M; Madabhushi, Anant; van der Laak, Jeroen; Ciompi, Francesco; Laenkholm, Anne-Vibeke; Bellolio, Enrique; Gruosso, Tina; Fox, Stephen B; Araya, Juan Carlos; Floris, Giuseppe; Hudeček, Jan; Voorwerk, Leonie; Beck, Andrew H; Kerner, Jen; Larsimont, Denis; Declercq, Sabine; Van den Eynden, Gert; Pusztai, Lajos; Ehinger, Anna; Yang, Wentao; AbdulJabbar, Khalid; Yuan, Yinyin; Singh, Rajendra; Hiley, Crispin; Bakir, Maise Al; Lazar, Alexander J; Naber, Stephen; Wienert, Stephan; Castillo, Miluska; Curigliano, Giuseppe; Dieci, Maria-Vittoria; André, Fabrice; Swanton, Charles; Reis-Filho, Jorge; Sparano, Joseph; Balslev, Eva; Chen, I-Chun; Stovgaard, Elisabeth Ida Specht; Pogue-Geile, Katherine; Blenman, Kim R M; Penault-Llorca, Frédérique; Schnitt, Stuart; Lakhani, Sunil R; Vincent-Salomon, Anne; Rojo, Federico; Braybrooke, Jeremy P; Hanna, Matthew G; Soler-Monsó, M Teresa; Bethmann, Daniel; Castaneda, Carlos A; Willard-Gallo, Karen; Sharma, Ashish; Lien, Huang-Chun; Fineberg, Susan; Thagaard, Jeppe; Comerma, Laura; Gonzalez-Ericsson, Paula; Brogi, Edi; Loi, Sherene; Saltz, Joel; Klaushen, Frederick; Cooper, Lee; Amgad, Mohamed; Moore, David A; Salgado, Roberto
Stromal tumor-infiltrating lymphocytes (sTILs) are important prognostic and predictive biomarkers in triple-negative (TNBC) and HER2-positive breast cancer. Incorporating sTILs into clinical practice necessitates reproducible assessment. Previously developed standardized scoring guidelines have been widely embraced by the clinical and research communities. We evaluated sources of variability in sTIL assessment by pathologists in three previous sTIL ring studies. We identify common challenges and evaluate impact of discrepancies on outcome estimates in early TNBC using a newly-developed prognostic tool. Discordant sTIL assessment is driven by heterogeneity in lymphocyte distribution. Additional factors include: technical slide-related issues; scoring outside the tumor boundary; tumors with minimal assessable stroma; including lymphocytes associated with other structures; and including other inflammatory cells. Small variations in sTIL assessment modestly alter risk estimation in early TNBC but have the potential to affect treatment selection if cutpoints are employed. Scoring and averaging multiple areas, as well as use of reference images, improve consistency of sTIL evaluation. Moreover, to assist in avoiding the pitfalls identified in this analysis, we developed an educational resource available at www.tilsinbreastcancer.org/pitfalls.
PMCID:7217863
PMID: 32411819
ISSN: 2374-4677
CID: 4431702

Application of a risk-management framework for integration of stromal tumor-infiltrating lymphocytes in clinical trials

Hudeček, Jan; Voorwerk, Leonie; van Seijen, Maartje; Nederlof, Iris; de Maaker, Michiel; van den Berg, Jose; van de Vijver, Koen K; Sikorska, Karolina; Adams, Sylvia; Demaria, Sandra; Viale, Giuseppe; Nielsen, Torsten O; Badve, Sunil S; Michiels, Stefan; Symmans, William Fraser; Sotiriou, Christos; Rimm, David L; Hewitt, Stephen M; Denkert, Carsten; Loibl, Sibylle; Loi, Sherene; Bartlett, John M S; Pruneri, Giancarlo; Dillon, Deborah A; Cheang, Maggie C U; Tutt, Andrew; Hall, Jacqueline A; Kos, Zuzana; Salgado, Roberto; Kok, Marleen; Horlings, Hugo M
Stromal tumor-infiltrating lymphocytes (sTILs) are a potential predictive biomarker for immunotherapy response in metastatic triple-negative breast cancer (TNBC). To incorporate sTILs into clinical trials and diagnostics, reliable assessment is essential. In this review, we propose a new concept, namely the implementation of a risk-management framework that enables the use of sTILs as a stratification factor in clinical trials. We present the design of a biomarker risk-mitigation workflow that can be applied to any biomarker incorporation in clinical trials. We demonstrate the implementation of this concept using sTILs as an integral biomarker in a single-center phase II immunotherapy trial for metastatic TNBC (TONIC trial, NCT02499367), using this workflow to mitigate risks of suboptimal inclusion of sTILs in this specific trial. In this review, we demonstrate that a web-based scoring platform can mitigate potential risk factors when including sTILs in clinical trials, and we argue that this framework can be applied for any future biomarker-driven clinical trial setting.
PMCID:7217941
PMID: 32436923
ISSN: 2374-4677
CID: 4444542