Try a new search

Format these results:

Searched for:

person:demars01

in-biosketch:yes

Total Results:

216


Radiation dose and fraction in immunotherapy: one-size regimen does not fit all settings, so how does one choose?

Demaria, Sandra; Guha, Chandan; Schoenfeld, Jonathan; Morris, Zachary; Monjazeb, Arta; Sikora, Andrew; Crittenden, Marka; Shiao, Stephen; Khleif, Samir; Gupta, Seema; Formenti, Silvia Chiara; Vikram, Bhadrasain; Coleman, C Norman; Ahmed, Mansoor M
Recent evidence indicates that ionizing radiation can enhance immune responses to tumors. Advances in radiation delivery techniques allow hypofractionated delivery of conformal radiotherapy. Hypofractionation or other modifications of standard fractionation may improve radiation's ability to promote immune responses to tumors. Other novel delivery options may also affect immune responses, including T-cell activation and tumor-antigen presentation changes. However, there is limited understanding of the immunological impact of hypofractionated and unique multifractionated radiotherapy regimens, as these observations are relatively recent. Hence, these differences in radiotherapy fractionation result in distinct immune-modulatory effects. Radiation oncologists and immunologists convened a virtual consensus discussion to identify current deficiencies, challenges, pitfalls and critical gaps when combining radiotherapy with immunotherapy and making recommendations to the field and advise National Cancer Institute on new directions and initiatives that will help further development of these two fields.This commentary aims to raise the awareness of this complexity so that the need to study radiation dose, fractionation, type and volume is understood and valued by the immuno-oncology research community. Divergence of approaches and findings between preclinical studies and clinical trials highlights the need for evaluating the design of future clinical studies with particular emphasis on radiation dose and fractionation, immune biomarkers and selecting appropriate end points for combination radiation/immune modulator trials, recognizing that direct effect on the tumor and potential abscopal effect may well be different. Similarly, preclinical studies should be designed as much as possible to model the intended clinical setting. This article describes a conceptual framework for testing different radiation therapy regimens as separate models of how radiation itself functions as an immunomodulatory 'drug' to provide alternatives to the widely adopted 'one-size-fits-all' strategy of frequently used 8 Gy×3 regimens immunomodulation.
PMID: 33827904
ISSN: 2051-1426
CID: 4839392

Differential and longitudinal immune gene patterns associated with reprogrammed microenvironment and viral mimicry in response to neoadjuvant radiotherapy in rectal cancer

Wilkins, Anna; Fontana, Elisa; Nyamundanda, Gift; Ragulan, Chanthirika; Patil, Yatish; Mansfield, David; Kingston, Jennifer; Errington-Mais, Fiona; Bottomley, Daniel; von Loga, Katharina; Bye, Hannah; Carter, Paul; Tinkler-Hundal, Emma; Noshirwani, Amir; Downs, Jessica; Dillon, Magnus; Demaria, Sandra; Sebag-Montefiore, David; Harrington, Kevin; West, Nick; Melcher, Alan; Sadanandam, Anguraj
BACKGROUND:Rectal cancers show a highly varied response to neoadjuvant radiotherapy/chemoradiation (RT/CRT) and the impact of the tumor immune microenvironment on this response is poorly understood. Current clinical tumor regression grading systems attempt to measure radiotherapy response but are subject to interobserver variation. An unbiased and unique histopathological quantification method (change in tumor cell density (ΔTCD)) may improve classification of RT/CRT response. Furthermore, immune gene expression profiling (GEP) may identify differences in expression levels of genes relevant to different radiotherapy responses: (1) at baseline between poor and good responders, and (2) longitudinally from preradiotherapy to postradiotherapy samples. Overall, this may inform novel therapeutic RT/CRT combination strategies in rectal cancer. METHODS:We generated GEPs for 53 patients from biopsies taken prior to preoperative radiotherapy. TCD was used to assess rectal tumor response to neoadjuvant RT/CRT and ΔTCD was subjected to k-means clustering to classify patients into different response categories. Differential gene expression analysis was performed using statistical analysis of microarrays, pathway enrichment analysis and immune cell type analysis using single sample gene set enrichment analysis. Immunohistochemistry was performed to validate specific results. The results were validated using 220 pretreatment samples from publicly available datasets at metalevel of pathway and survival analyses. RESULTS:ΔTCD scores ranged from 12.4% to -47.7% and stratified patients into three response categories. At baseline, 40 genes were significantly upregulated in poor (n=12) versus good responders (n=21), including myeloid and stromal cell genes. Of several pathways showing significant enrichment at baseline in poor responders, epithelial to mesenchymal transition, coagulation, complement activation and apical junction pathways were validated in external cohorts. Unlike poor responders, good responders showed longitudinal (preradiotherapy vs postradiotherapy samples) upregulation of 198 immune genes, reflecting an increased T-cell-inflamed GEP, type-I interferon and macrophage populations. Longitudinal pathway analysis suggested viral-like pathogen responses occurred in post-treatment resected samples compared with pretreatment biopsies in good responders. CONCLUSION/CONCLUSIONS:This study suggests potentially druggable immune targets in poor responders at baseline and indicates that tumors with a good RT/CRT response reprogrammed from immune "cold" towards an immunologically "hot" phenotype on treatment with radiotherapy.
PMID: 33678606
ISSN: 2051-1426
CID: 4819602

Preventive efficacy of a tenofovir alafenamide fumarate nanofluidic implant in SHIV-challenged nonhuman primates

Pons-Faudoa, Fernanda P; Sizovs, Antons; Shelton, Kathryn A; Momin, Zoha; Niles, Jean A; Bushman, Lane R; Xu, Jiaqiong; Chua, Corrine Ying Xuan; Nichols, Joan E; Demaria, Sandra; Ittmann, Michael M; Hawkins, Trevor; Rooney, James F; Marzinke, Mark A; Kimata, Jason T; Anderson, Peter L; Nehete, Pramod N; Arduino, Roberto C; Ferrari, Mauro; Sastry, K Jagannadha; Grattoni, Alessandro
Pre-exposure prophylaxis (PrEP) using antiretroviral oral drugs is effective at preventing HIV transmission when individuals adhere to the dosing regimen. Tenofovir alafenamide (TAF) is a potent antiretroviral drug, with numerous long-acting (LA) delivery systems under development to improve PrEP adherence. However, none has undergone preventive efficacy assessment. Here we show that LA TAF using a novel subcutaneous nanofluidic implant (nTAF) confers partial protection from HIV transmission. We demonstrate that sustained subcutaneous delivery through nTAF in rhesus macaques maintained tenofovir diphosphate concentration at a median of 390.00 fmol/106 peripheral blood mononuclear cells, 9 times above clinically protective levels. In a non-blinded, placebo-controlled rhesus macaque study with repeated low-dose rectal SHIVSF162P3 challenge, the nTAF cohort had a 62.50% reduction (95% CI: 1.72% to 85.69%; p=0.068) in risk of infection per exposure compared to the control. Our finding mirrors that of tenofovir disoproxil fumarate (TDF) monotherapy, where 60.00% protective efficacy was observed in macaques, and clinically, 67.00% reduction in risk with 86.00% preventive efficacy in individuals with detectable drug in the plasma. Overall, our nanofluidic technology shows potential as a subcutaneous delivery platform for long-term PrEP and provides insights for clinical implementation of LA TAF for HIV prevention.
PMCID:8114879
PMID: 33997267
ISSN: 2366-3987
CID: 4876592

Activin A promotes regulatory T cell-mediated immunosuppression in irradiated breast cancer

De Martino, Mara; Daviaud, Camille; Diamond, Julie M; Kraynak, Jeffrey; Alard, Amandine; Formenti, Silvia C; Miller, Lance D; Demaria, Sandra; Vanpouille-Box, Claire
Increased regulatory T cells (Tregs) after radiation therapy have been reported, but the mechanisms of their induction remain incompletely understood. TGFβ is known to foster Treg differentiation within tumors and is activated following radiation therapy. Thus, we hypothesized that TGFβ blockade would result in decreased Tregs within the irradiated tumor microenvironment (TME). We found increased Tregs in the tumors of mice treated with focal radiotherapy and TGFβ blockade. This increase was mediated by upregulation of another TGFβ family member, activin A. In vitro, activin A secretion was increased following irradiation of mouse and human breast cancer cells, and its expression was further enhanced upon TGFβ blockade. In vivo, dual blockade of activin A and TGFβ was required to decrease intratumoral Tregs in the context of radiation. This resulted in an increase in CD8+ T-cell priming and was associated with a reduced tumor recurrence rate. Combination of immune checkpoint inhibitors with the dual blockade of activin A and TGFβ led to the development of tumor-specific memory responses in irradiated breast cancer. Supporting the translational value of activin A targeting to reduce Treg-mediated immunosuppression, retrospective analysis of a public dataset of breast cancer patients revealed a positive correlation between activin A gene expression and Treg abundance. Overall, these results shed light on an immune escape mechanism driven by activin A, and suggest that dual targeting of activin A and TGFβ may be required to optimally unleash radiation-induced antitumor immunity against breast cancer.
PMID: 33093219
ISSN: 2326-6074
CID: 4642872

Radiation-induced adaptive response: new potential for cancer treatment

Coleman, C Norman; Eke, Iris; Makinde, Adeola Y; Chopra, Sunita; Demaria, Sandra; Formenti, Silvia C; Martello, Shannon; Bylicky, Michelle; Mitchell, James B; Aryankalayil, Molykutty J
Radiation therapy (RT) is highly effective due to its ability to physically focus the treatment to target the tumor while sparing normal tissue and its ability to be combined with systemic therapy. This systemic therapy can be utilized before RT as an adjuvant or induction treatment, during RT as a radiation "sensitizer," or following RT as a part of combined modality therapy. As part of a unique concept of using radiation as "focused biology" we investigated how tumors and normal tissues adapt to clinically relevant multi-fraction (MF) and single-dose (SD) radiation to observe whether the adaptations can induce susceptibility to cell killing by available drugs or by immune enhancement. We identified an adaptation occurring after MF (3 x 2 Gy) that induced cell killing when AKT-mTOR inhibitors were delivered following cessation of RT. Additionally, we identified inducible changes in integrin expression 2 months following cessation of RT that differ between MF (1 Gy x 10) and SD (10 Gy) that remain targetable compared to pre-RT. Adaptation is reflected across different "omics" studies, and thus the range of possible molecular targets is not only broad but also time, dose, and schedule dependent. While much remains to be studied about the radiation adaptive response, radiation should be characterized by its molecular perturbations in addition to physical dose. Consideration of the adaptive effects should result in the design of a tailored radiotherapy treatment plan that accounts for specific molecular changes to be targeted as part of precision multi-modality cancer treatment.
PMID: 32554542
ISSN: 1078-0432
CID: 4485092

Converging focal radiation and immunotherapy in a preclinical model of triple negative breast cancer: contribution of VISTA blockade

Pilones, Karsten A; Hensler, Michal; Daviaud, Camille; Kraynak, Jeffrey; Fucikova, Jitka; Galluzzi, Lorenzo; Demaria, Sandra; Formenti, Silvia C
Antibodies targeting the co-inhibitory receptor programmed cell death 1 (PDCD1, best known as PD-1) or its main ligand CD274 (best known as PD-L1) have shown some activity in patients with metastatic triple-negative breast cancer (TNBC), especially in a recent Phase III clinical trial combining PD-L1 blockade with taxane-based chemotherapy. Despite these encouraging findings, however, most patients with TNBC fail to derive significant benefits from PD-L1 blockade, calling for the identification of novel therapeutic approaches. Here, we used the 4T1 murine mammary cancer model of metastatic and immune-resistant TNBC to test whether focal radiation therapy (RT), a powerful inducer of immunogenic cell death, in combination with various immunotherapeutic strategies can overcome resistance to immune checkpoint blockade. Our results suggest that focal RT enhances the therapeutic effects of PD-1 blockade against primary 4T1 tumors and their metastases. Similarly, the efficacy of an antibody specific for V-set immunoregulatory receptor (VSIR, another co-inhibitory receptor best known as VISTA) was enhanced by focal RT. Administration of cyclophosphamide plus RT and dual PD-1/VISTA blockade had superior therapeutic effects, which were associated with activation of tumor-infiltrating CD8+ T cells and depletion of intratumoral granulocytic myeloid-derived suppressor cells (MDSCs). Overall, these results demonstrate that RT can sensitize immunorefractory tumors to VISTA or PD-1 blockade, that this effect is enhanced by the addition of cyclophosphamide and suggest that a multipronged immunotherapeutic approach may also be required to increase the incidence of durable responses in patients with TNBC.
PMCID:7583495
PMID: 33150045
ISSN: 2162-4011
CID: 4661562

Perspectives in melanoma: meeting report from the "Melanoma Bridge" (December 5th-7th, 2019, Naples, Italy)

Ascierto, Paolo A; Puzanov, Igor; Agarwala, Sanjiv S; Blank, Christian; Carvajal, Richard D; Demaria, Sandra; Dummer, Reinhard; Ernstoff, Marc; Ferrone, Soldano; Fox, Bernard A; Gajewski, Thomas F; Garbe, Claus; Hwu, Patrick; Lo, Roger S; Long, Georgina V; Luke, Jason J; Osman, Iman; Postow, Michael A; Sullivan, Ryan J; Taube, Janis M; Trinchieri, Giorgio; Zarour, Hassane M; Caracò, Corrado; Thurin, Magdalena
The melanoma treatment landscape changed in 2011 with the approval of the first anti-cytotoxic T-lymphocyte-associated protein (CTLA)-4 checkpoint inhibitor and of the first BRAF-targeted monoclonal antibody, both of which significantly improved overall survival (OS). Since then, improved understanding of the tumor microenvironment (TME) and tumor immune-evasion mechanisms has resulted in new approaches to targeting and harnessing the host immune response. The approval of new immune and targeted therapies has further improved outcomes for patients with advanced melanoma and other combination modalities are also being explored such as chemotherapy, radiotherapy, electrochemotherapy and surgery. In addition, different strategies of drugs administration including sequential or combination treatment are being tested. Approaches to overcome resistance and to potentiate the immune response are being developed. Increasing evidence emerges that tissue and blood-based biomarkers can predict the response to a therapy. The latest findings in melanoma research, including insights into the tumor microenvironment and new biomarkers, improved understanding of tumor immune response and resistance, novel approaches for combination strategies and the role of neoadjuvant and adjuvant therapy, were the focus of discussions at the Melanoma Bridge meeting (5-7 December, 2019, Naples, Italy), which are summarized in this report.
PMID: 32894202
ISSN: 1479-5876
CID: 4588772

Future of Radiation and Immunotherapy [Editorial]

Formenti, Silvia C; Demaria, Sandra
PMID: 32819614
ISSN: 1879-355x
CID: 4567322

Radiotherapy cooperates with IL15 to induce antitumor immune responses

Pilones, Karsten A; Charpentier, Maud; Garcia-Martinez, Elena; Daviaud, Camille; Kraynak, Jeffrey; Aryankalayil, Joseph; Formenti, Silvia C; Demaria, Sandra
Focal radiotherapy can promote cross-presentation of tumor antigens to T cells, but by itself it is insufficient to induce therapeutically effective T-cell responses. The common gamma-chain cytokine IL15 promotes and sustains the proliferation and effector function of CD8+ T cells, but has limited activity against poorly immunogenic tumors that do not elicit significant spontaneous T-cell responses. Here, we show that radiotherapy and subcutaneous IL15 had complementary effects and induced CD8+ T cell-mediated tumor regression and long-term protective memory responses in two mouse carcinoma models unresponsive to IL15 alone. Mechanistically, radiotherapy-induced IFN type I production and Batf3-dependent conventional dendritic cells type 1 (cDC1s) were required for priming of tumor-specific CD8+ T cells and for the therapeutic effect of the combination. IL15 cooperated with radiotherapy to activate and recruit cDC1s to the tumor. IL15 alone and in complex with a hybrid molecule containing the IL15α receptor have been tested in early phase clinical trials in cancer patients and demonstrated good tolerability, especially when given subcutaneously. Expansion of NK cells and CD8+ T cells were noted, without clear clinical activity, suggesting further testing of IL15 as a component of a combinatorial treatment with other agents. Our results provide the rationale for testing combinations of IL15 with radiotherapy in the clinic.
PMID: 32532811
ISSN: 2326-6074
CID: 4482062

IL15 synergizes with radiotherapy to reprogram the tumor immune contexture through a dendritic cell connection

Pilones, Karsten A; Charpentier, Maud; Garcia-Martinez, Elena; Demaria, Sandra
IL15 is a key cytokine for the activation and survival of anti-tumor effectors CD8+ T and NK cells. Recently published preclinical studies demonstrate that the therapeutic activity of IL15 requires conventional dendritic cells type 1 (cDC1). Radiotherapy cooperates with IL15 by enhancing cDC1 tumor infiltration via interferon type 1 activation.
PMCID:7466855
PMID: 32934886
ISSN: 2162-4011
CID: 4593022