Try a new search

Format these results:

Searched for:

person:formes01

Total Results:

488


PD-1 blockade in recurrent or metastatic cervical cancer: Data from cemiplimab phase I expansion cohorts and characterization of PD-L1 expression in cervical cancer

Rischin, Danny; Gil-Martin, Marta; González-Martin, Antonio; Braña, Irene; Hou, June Y; Cho, Daniel; Falchook, Gerald S; Formenti, Silvia; Jabbour, Salma; Moore, Kathleen; Naing, Aung; Papadopoulos, Kyriakos P; Baranda, Joaquina; Fury, Wen; Feng, Minjie; Stankevich, Elizabeth; Li, Jingjin; Yama-Dang, N Alice; Yoo, Suk-Young; Lowy, Israel; Mathias, Melissa; Fury, Matthew G
OBJECTIVES/OBJECTIVE:To characterize the safety, tolerability, and anti-tumor activity of cemiplimab as monotherapy or in combination with hypofractionated radiation therapy (hfRT) in patients with recurrent or metastatic cervical cancer. To determine the association between histology and programmed death-ligand 1 (PD-L1) expression. METHODS:In non-randomized phase I expansion cohorts, patients (squamous or non-squamous histology) received cemiplimab 3 mg/kg intravenously every 2 weeks for 48 weeks, either alone (monotherapy cohort) or with hfRT during week 2 (combination cohort). Due to insufficient tissue material, PD-L1 protein expression was evaluated in commercially purchased samples and mRNA expression levels were analyzed from The Cancer Genome Atlas (TCGA). RESULTS:Twenty patients enrolled in both cohorts in total; 10 had squamous histology. The most common adverse events of any grade were diarrhea, fatigue, and hypokalemia, occurring in 35%, 25%, and 25%, respectively. Objective response rate was 10% in each cohort; responders had squamous histology. Duration of response was 11.2 months and 6.4 months for the responder in the monotherapy and combination cohort, respectively. Irradiated lesions were not included in the response assessments. In separate archived specimens (N = 155), PD-L1 protein expression in tumor and immune cells was negative (<1%) more commonly in adenocarcinoma than in squamous tumors. PD-L1 mRNA levels were lower in adenocarcinoma than squamous cell tumors (1.2 vs 5.0 mean transcripts per million, respectively) in TCGA. CONCLUSIONS:Cemiplimab has activity in cervical squamous cell carcinoma. The phase I results, combined with results from other anti-PD-1 trials in cervical cancer and our biomarker analyses have informed the design of the ongoing phase III trial, with the primary overall survival hierarchical analyses being done first in patients with squamous histology.
PMID: 32917410
ISSN: 1095-6859
CID: 4615542

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

Mitochondrial DNA drives abscopal responses to radiation that are inhibited by autophagy

Yamazaki, Takahiro; Kirchmair, Alexander; Sato, Ai; Buqué, Aitziber; Rybstein, Marissa; Petroni, Giulia; Bloy, Norma; Finotello, Francesca; Stafford, Lena; Navarro Manzano, Esther; Ayala de la Peña, Francisco; García-Martínez, Elena; Formenti, Silvia C; Trajanoski, Zlatko; Galluzzi, Lorenzo
Autophagy supports both cellular and organismal homeostasis. However, whether autophagy should be inhibited or activated for cancer therapy remains unclear. Deletion of essential autophagy genes increased the sensitivity of mouse mammary carcinoma cells to radiation therapy in vitro and in vivo (in immunocompetent syngeneic hosts). Autophagy-deficient cells secreted increased amounts of type I interferon (IFN), which could be limited by CGAS or STING knockdown, mitochondrial DNA depletion or mitochondrial outer membrane permeabilization blockage via BCL2 overexpression or BAX deletion. In vivo, irradiated autophagy-incompetent mammary tumors elicited robust immunity, leading to improved control of distant nonirradiated lesions via systemic type I IFN signaling. Finally, a genetic signature of autophagy had negative prognostic value in patients with breast cancer, inversely correlating with mitochondrial abundance, type I IFN signaling and effector immunity. As clinically useful autophagy inhibitors are elusive, our findings suggest that mitochondrial outer membrane permeabilization may represent a valid target for boosting radiation therapy immunogenicity in patients with breast cancer.
PMID: 32747819
ISSN: 1529-2916
CID: 4583722

Publisher Correction: Immunoprophylactic and immunotherapeutic control of hormone receptor-positive breast cancer

Buqué, Aitziber; Bloy, Norma; Perez-Lanzón, Maria; Iribarren, Kristina; Humeau, Juliette; Pol, Jonathan G; Levesque, Sarah; Mondragon, Laura; Yamazaki, Takahiro; Sato, Ai; Aranda, Fernando; Durand, Sylvère; Boissonnas, Alexandre; Fucikova, Jitka; Senovilla, Laura; Enot, David; Hensler, Michal; Kremer, Margerie; Stoll, Gautier; Hu, Yang; Massa, Chiara; Formenti, Silvia C; Seliger, Barbara; Elemento, Olivier; Spisek, Radek; André, Fabrice; Zitvogel, Laurence; Delaloge, Suzette; Kroemer, Guido; Galluzzi, Lorenzo
An amendment to this paper has been published and can be accessed via a link at the top of the paper.
PMID: 32943636
ISSN: 2041-1723
CID: 4627492

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

Immunoprophylactic and immunotherapeutic control of hormone receptor-positive breast cancer

Buqué, Aitziber; Bloy, Norma; Perez-Lanzón, Maria; Iribarren, Kristina; Humeau, Juliette; Pol, Jonathan G; Levesque, Sarah; Mondragon, Laura; Yamazaki, Takahiro; Sato, Ai; Aranda, Fernando; Durand, Sylvère; Boissonnas, Alexandre; Fucikova, Jitka; Senovilla, Laura; Enot, David; Hensler, Michal; Kremer, Margerie; Stoll, Gautier; Hu, Yang; Massa, Chiara; Formenti, Silvia C; Seliger, Barbara; Elemento, Olivier; Spisek, Radek; André, Fabrice; Zitvogel, Laurence; Delaloge, Suzette; Kroemer, Guido; Galluzzi, Lorenzo
Hormone receptor (HR)+ breast cancer (BC) causes most BC-related deaths, calling for improved therapeutic approaches. Despite expectations, immune checkpoint blockers (ICBs) are poorly active in patients with HR+ BC, in part reflecting the lack of preclinical models that recapitulate disease progression in immunocompetent hosts. We demonstrate that mammary tumors driven by medroxyprogesterone acetate (M) and 7,12-dimethylbenz[a]anthracene (D) recapitulate several key features of human luminal B HR+HER2- BC, including limited immune infiltration and poor sensitivity to ICBs. M/D-driven oncogenesis is accelerated by immune defects, demonstrating that M/D-driven tumors are under immunosurveillance. Safe nutritional measures including nicotinamide (NAM) supplementation efficiently delay M/D-driven oncogenesis by reactivating immunosurveillance. NAM also mediates immunotherapeutic effects against established M/D-driven and transplantable BC, largely reflecting increased type I interferon secretion by malignant cells and direct stimulation of immune effector cells. Our findings identify NAM as a potential strategy for the prevention and treatment of HR+ BC.
PMID: 32732875
ISSN: 2041-1723
CID: 4542902

Understanding High-Dose, Ultra-High Dose Rate, and Spatially Fractionated Radiation Therapy

Griffin, Robert J; Ahmed, Mansoor M; Amendola, Beatriz; Belyakov, Oleg; Bentzen, Søren M; Butterworth, Karl T; Chang, Sha; Coleman, C Norman; Djonov, Valentin; Formenti, Sylvia C; Glatstein, Eli; Guha, Chandan; Kalnicki, Shalom; Le, Quynh-Thu; Loo, Billy W; Mahadevan, Anand; Massaccesi, Mariangela; Maxim, Peter G; Mohiuddin, Majid; Mohiuddin, Mohammed; Mayr, Nina A; Obcemea, Ceferino; Petersson, Kristoffer; Regine, William; Roach, Mack; Romanelli, Pantaleo; Simone, Charles B; Snider, James W; Spitz, Douglas R; Vikram, Bhadrasain; Vozenin, Marie-Catherine; Abdel-Wahab, May; Welsh, James; Wu, Xiaodong; Limoli, Charles L
The National Cancer Institute's Radiation Research Program, in collaboration with the Radiosurgery Society, hosted a workshop called Understanding High-Dose, Ultra-High Dose Rate and Spatially Fractionated Radiotherapy on August 20 and 21, 2018 to bring together experts in experimental and clinical experience in these and related fields. Critically, the overall aims were to understand the biological underpinning of these emerging techniques and the technical/physical parameters that must be further defined to drive clinical practice through innovative biologically based clinical trials.
PMID: 32298811
ISSN: 1879-355x
CID: 5194772

Cancer and COVID-19 - potentially deleterious effects of delaying radiotherapy

Nagar, Himanshu; Formenti, Silvia C
PMCID:7184542
PMID: 32341524
ISSN: 1759-4782
CID: 4505002

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