Searched for: person:formes01
Current clinical trials testing the combination of immunotherapy with radiotherapy
Kang, Josephine; Demaria, Sandra; Formenti, Silvia
Increasing evidence demonstrates that radiation acts as an immune stimulus, recruiting immune mediators that enable anti-tumor responses within and outside the radiation field. There has been a rapid expansion in the number of clinical trials harnessing radiation to enhance antitumor immunity. If positive, results of these trials will lead to a paradigm shift in the use of radiotherapy. In this review, we discuss the rationale for trials combining radiation with various immunotherapies, provide an update of recent clinical trial results and highlight trials currently in progress. We also address issues pertaining to the optimal incorporation of immunotherapy with radiation, including sequencing of treatment, radiation dosing and evaluation of clinical trial endpoints.
PMCID:5028964
PMID: 27660705
ISSN: 2051-1426
CID: 3091202
In situ vaccination by radiotherapy to improve responses to anti-CTLA-4 treatment
Vanpouille-Box, Claire; Pilones, Karsten A; Wennerberg, Erik; Formenti, Silvia C; Demaria, Sandra
Targeting immune checkpoint receptors has emerged as an effective strategy to induce immune-mediated cancer regression in the subset of patients who have significant pre-existing anti-tumor immunity. For the remainder, effective anti tumor responses may require vaccination. Radiotherapy, traditionally used to achieve local tumor control, has acquired a new role, that of a partner for immunotherapy. Ionizing radiation has pro-inflammatory effects that facilitate tumor rejection. Radiation alters the tumor to enhance the concentration of effector T cells via induction of chemokines, cytokines and adhesion molecules. In parallel, radiation can induce an immunogenic death of cancer cells, promoting cross-presentation of tumor-derived antigens by dendritic cells to T cells. Newly generated anti-tumor immune responses have been demonstrated post-radiation in both murine models and occasional patients, supporting the hypothesis that the irradiated tumor can become an in situ vaccine. It is in this role, that radiation can be applied to induce anti-tumor T cells in lymphocyte-poor tumors, and possibly benefit patients who would otherwise fail to respond to immune checkpoint inhibitors. This review summarizes preclinical and clinical data demonstrating that radiation acts in concert with antibodies targeting the immune checkpoint cytotoxic T-lymphocyte antigen-4 (CTLA-4), to induce therapeutically effective anti-tumor T cell responses in tumors otherwise non responsive to anti-CTLA-4 therapy.
PMCID:4684480
PMID: 26148880
ISSN: 1873-2518
CID: 1662622
Mechanisms of synergy of radiotherapy and immunotherapy [Meeting Abstract]
Pilones, K A; Emerson, R; Formenti, S C; Robbins, H S; Demaria, S
The success of immune checkpoint inhibitors in inducing tumor regression has demonstrated that specific inhibitory pathways are dominant rate-limiting steps in a significant number of patients with melanoma and other advanced cancers. However, in the majority of patients tumor rejection is hindered by multiple immunosuppressive mechanisms present in the tumor microenvironment. Obstacles to immune-mediated tumor control can be present at both the priming and effector phase of the anti-tumor response, and include defective function and activation of antigen-presenting cells, defective T cell recruitment and infiltration of tumors, and defective recognition and killing of cancer cells by T cells. Ionizing radiation therapy (RT) applied locally to a tumor at therapeutic doses has multiple effects that can potentially overcome each of these obstacles, and we have shown that RT is synergistic with immunotherapy. In pre-clinical models RT converted tumors unresponsive to anti-CTLA-4 mAb into responsive ones, achieving rejection of the irradiated tumor and non-irradiated metastases (abscopal effect) and improved survival [1,2]. At the effector phase, RT enhanced recruitment of activated T cells to the tumor by induction of chemokines [3], and enhanced immune synapse formation between CD8 T and tumor cells by induction of NKG2D ligands [4]. To test the hypothesis that successful tumor rejection induced by RT +anti-CTLA-4 requires a significant change in the quantity and quality of tumor-infiltrating lymphocytes (TILs) we performed a comprehensive evaluation of the breadth and depth of the T cell repertoire modulated by treatment using high-throughput sequencing technology. To gain insights into the changes in TILs induced by anti-CTLA-4 treatment in tumor hosts that respond or do not respond to therapy we used our well-characterized 4T1 mouse model in which anti-CTLA-4 treatment is effective only when combined with RT. Results show distinct contributions of RT and anti-CTLA-4 to increasing the number and clonality of TILs, and changes in clonal representation that are unique to the combination. These data suggest that RT effectively releases endogenous tumor antigens that prime anti-tumor T cells, supporting the concept that it can be used as a mean to generate an in situ individualized vaccine. We are currently exploring this hypothesis in clinical trials testing the combination of RT and checkpoint inhibitors
EMBASE:72289909
ISSN: 1479-5876
CID: 2150452
Cesium-131 brachytherapy in high risk and recurrent head and neck cancers: first report of long-term outcomes
Pham, Anthony; Arora, Shruthi; Wernicke, A Gabriella; Kutler, David I; Cohen, Marc; Kuhel, William; Trichter, Samuel; Nori, Dattatreyudu; Formenti, Silvia C; Parashar, Bhupesh
PURPOSE: The feasibility and efficacy of re-irradiation using contemporary radiation techniques to treat recurrent head and neck cancer has been demonstrated but the role of brachytherapy is unclear. Here we describe the use of (131)Cs brachytherapy with concurrent salvage surgery in 18 patients. MATERIAL AND METHODS: Eligible patients underwent maximal gross resection of the tumor with implantation of brachytherapy seeds delivering a minimum dose of 80 Gy to the tumor bed. Rates of overall survival, locoregional progression free survival, disease-free survival, and radiation-induced toxicity were analyzed. RESULTS: Retrospective Kaplan-Meier analysis shows median overall survival was 15 months and disease free survival was 12 months. Two patients developed grade 3 toxicity; all other complications were grade 1-2 with no grade 4 or 5 complications. CONCLUSIONS: Compared to prior literature, our study shows comparable rates of survival with a decreased rate of radiation-induced toxicity.
PMCID:4716131
PMID: 26816501
ISSN: 1689-832x
CID: 2043962
Role of Local Radiation Therapy in Cancer Immunotherapy
Demaria, Sandra; Golden, Encouse B; Formenti, Silvia C
The recent success of cancer immunotherapy has demonstrated the power of the immune system to clear tumors, generating renewed enthusiasm for identifying ways to induce antitumor immune responses in patients. Natural antitumor immune responses are detectable in a fraction of patients across multiple malignant neoplasms and can be reactivated by targeting rate-limiting immunosuppressive mechanisms. In most patients, however, interventions to induce a de novo antitumor immune response are necessary. We review growing evidence that radiation therapy targeted to the tumor can convert it into an in situ tumor vaccine by inducing release of antigens during cancer cell death in association with proinflammatory signals that trigger the innate immune system to activate tumor-specific T cells. In addition, radiation's effects on the tumor microenvironment enhance infiltration of activated T cells and can overcome some of the barriers to tumor rejection. Thus, the complementary effects of radiation on priming and effector phases of antitumor immunity make it an appealing strategy to generate immunity against a patient's own individual tumor, that through immunological memory, can result in long-lasting systemic responses. Several anecdotal cases have demonstrated the efficacy of combining radiation with available immunotherapies, and results of prospective trials are forthcoming.
PMID: 26270858
ISSN: 2374-2445
CID: 1721812
Future perspectives in melanoma research: meeting report from the "Melanoma Bridge": Napoli, December 3rd-6th 2014
Ascierto, Paolo A; Atkins, Michael; Bifulco, Carlo; Botti, Gerardo; Cochran, Alistair; Davies, Michael; Demaria, Sandra; Dummer, Reinhard; Ferrone, Soldano; Formenti, Silvia; Gajewski, Thomas F; Garbe, Claus; Khleif, Samir; Kiessling, Rolf; Lo, Roger; Lorigan, Paul; Arthur, Grant Mc; Masucci, Giuseppe; Melero, Ignacio; Mihm, Martin; Palmieri, Giuseppe; Parmiani, Giorgio; Puzanov, Igor; Romero, Pedro; Schilling, Bastian; Seliger, Barbara; Stroncek, David; Taube, Janis; Tomei, Sara; Zarour, Hassane M; Testori, Alessandro; Wang, Ena; Galon, Jerome; Ciliberto, Gennaro; Mozzillo, Nicola; Marincola, Francesco M; Thurin, Magdalena
The fourth "Melanoma Bridge Meeting" took place in Naples, December 3-6th, 2014. The four topics discussed at this meeting were: Molecular and Immunological Advances, Combination Therapies, News in Immunotherapy, and Tumor Microenvironment and Biomarkers. Until recently systemic therapy for metastatic melanoma patients was ineffective, but recent advances in tumor biology and immunology have led to the development of new targeted and immunotherapeutic agents that prolong progression-free survival (PFS) and overall survival (OS). New therapies, such as mitogen-activated protein kinase (MAPK) pathway inhibitors as well as other signaling pathway inhibitors, are being tested in patients with metastatic melanoma either as monotherapy or in combination, and all have yielded promising results. These include inhibitors of receptor tyrosine kinases (BRAF, MEK, and VEGFR), the phosphatidylinositol 3 kinase (PI3K) pathway [PI3K, AKT, mammalian target of rapamycin (mTOR)], activators of apoptotic pathway, and the cell cycle inhibitors (CDK4/6). Various locoregional interventions including radiotherapy and surgery are still valid approaches in treatment of advanced melanoma that can be integrated with novel therapies. Intrinsic, adaptive and acquired resistance occur with targeted therapy such as BRAF inhibitors, where most responses are short-lived. Given that the reactivation of the MAPK pathway through several distinct mechanisms is responsible for the majority of acquired resistance, it is logical to combine BRAF inhibitors with inhibitors of targets downstream in the MAPK pathway. For example, combination of BRAF/MEK inhibitors (e.g., dabrafenib/trametinib) have been demonstrated to improve survival compared to monotherapy. Application of novel technologies such sequencing have proven useful as a tool for identification of MAPK pathway-alternative resistance mechanism and designing other combinatorial therapies such as those between BRAF and AKT inhibitors. Improved survival rates have also been observed with immune-targeted therapy for patients with metastatic melanoma. Immune-modulating antibodies came to the forefront with anti-CTLA-4, programmed cell death-1 (PD-1) and PD-1 ligand 1 (PD-L1) pathway blocking antibodies that result in durable responses in a subset of melanoma patients. Agents targeting other immune inhibitory (e.g., Tim-3) or immune stimulating (e.g., CD137) receptors and other approaches such as adoptive cell transfer demonstrate clinical benefit in patients with melanoma as well. These agents are being studied in combination with targeted therapies in attempt to produce longer-term responses than those more typically seen with targeted therapy. Other combinations with cytotoxic chemotherapy and inhibitors of angiogenesis are changing the evolving landscape of therapeutic options and are being evaluated to prevent or delay resistance and to further improve survival rates for this patient population. This meeting's specific focus was on advances in combination of targeted therapy and immunotherapy. Both combination targeted therapy approaches and different immunotherapies were discussed. Similarly to the previous meetings, the importance of biomarkers for clinical application as markers for diagnosis, prognosis and prediction of treatment response was an integral part of the meeting. The overall emphasis on biomarkers supports novel concepts toward integrating biomarkers into contemporary clinical management of patients with melanoma across the entire spectrum of disease stage. Translation of the knowledge gained from the biology of tumor microenvironment across different tumors represents a bridge to impact on prognosis and response to therapy in melanoma.
PMCID:4665874
PMID: 26619946
ISSN: 1479-5876
CID: 2040952
Anticancer immunotherapy by CTLA-4 blockade relies on the gut microbiota
Vetizou, Marie; Pitt, Jonathan M; Daillere, Romain; Lepage, Patricia; Waldschmitt, Nadine; Flament, Caroline; Rusakiewicz, Sylvie; Routy, Bertrand; Roberti, Maria P; Duong, Connie P M; Poirier-Colame, Vichnou; Roux, Antoine; Becharef, Sonia; Formenti, Silvia; Golden, Encouse; Cording, Sascha; Eberl, Gerard; Schlitzer, Andreas; Ginhoux, Florent; Mani, Sridhar; Yamazaki, Takahiro; Jacquelot, Nicolas; Enot, David P; Berard, Marion; Nigou, Jerome; Opolon, Paule; Eggermont, Alexander; Woerther, Paul-Louis; Chachaty, Elisabeth; Chaput, Nathalie; Robert, Caroline; Mateus, Christina; Kroemer, Guido; Raoult, Didier; Boneca, Ivo Gomperts; Carbonnel, Franck; Chamaillard, Mathias; Zitvogel, Laurence
Antibodies targeting CTLA-4 have been successfully used as cancer immunotherapy. We find that the antitumor effects of CTLA-4 blockade depend on distinct Bacteroides species. In mice and patients, T cell responses specific for B. thetaiotaomicron or B. fragilis were associated with the efficacy of CTLA-4 blockade. Tumors in antibiotic-treated or germ-free mice did not respond to CTLA blockade. This defect was overcome by gavage with B. fragilis, by immunization with B. fragilis polysaccharides, or by adoptive transfer of B. fragilis-specific T cells. Fecal microbial transplantation from humans to mice confirmed that treatment of melanoma patients with antibodies against CTLA-4 favored the outgrowth of B. fragilis with anticancer properties. This study reveals a key role for Bacteroidales in the immunostimulatory effects of CTLA-4 blockade.
PMCID:4721659
PMID: 26541610
ISSN: 1095-9203
CID: 2210722
Optimal Topical Agent for Radiation Dermatitis During Breast Radiotherapy: A Pilot Study
Fenton-Kerimian, Maria; Cartwright, Frances; Peat, Elicia; Florentino, Rosanna; Maisonet, Olivier; Budin, Wendy; Rolnitzky, Linda; Formenti, Silvia
BACKGROUND: Women receiving radiation to the breast will likely be recommended to use a topical cream to minimize and delay the development of radiation dermatitis. Although many topical products are commercially available and have been tested for safety and efficacy, few studies have compared various products to one another for superiority and cost effectiveness. OBJECTIVES: The purpose of this pilot study was to compare three commonly used skin care products prospectively to one other in a homogenously controlled group of women undergoing whole breast irradiation to assess superiority in minimizing the common toxicity criteria grade of radiation dermatitis, effect on quality of life, and cost. METHODS: The authors conducted a systematic review to determine the three types of skin care products with the strongest evidence of minimizing radiation dermatitis. Patients were voluntarily enrolled and randomized to one of three possible skin care topical regimens. Patients completed a quality-of-life survey to assess their preference in topical skin care regimen. The cost of each arm's topical product was assessed at the completion of patient participation. FINDINGS: No statistical difference was noted in the severity or occurrence of radiation dermatitis among the groups. In addition, no statistical difference was found among the three treatment arms in quality-of-life score changes, and no patients required a treatment interruption in their radiation or in the skin care product during treatment. A cost difference among the treatment arms was noted.
PMID: 26207710
ISSN: 1538-067x
CID: 1684162
Local radiotherapy and granulocyte-macrophage colony-stimulating factor to generate abscopal responses in patients with metastatic solid tumours: a proof-of-principle trial
Golden, Encouse B; Chhabra, Arpit; Chachoua, Abraham; Adams, Sylvia; Donach, Martin; Fenton-Kerimian, Maria; Friedman, Kent; Ponzo, Fabio; Babb, James S; Goldberg, Judith; Demaria, Sandra; Formenti, Silvia C
BACKGROUND: An abscopal response describes radiotherapy-induced immune-mediated tumour regression at sites distant to the irradiated field. Granulocyte-macrophage colony-stimulating factor is a potent stimulator of dendritic cell maturation. We postulated that the exploitation of the pro-immunogenic effects of radiotherapy with granulocyte-macrophage colony-stimulating factor might result in abscopal responses among patients with metastatic cancer. METHODS: Patients with stable or progressing metastatic solid tumours, on single-agent chemotherapy or hormonal therapy, with at least three distinct measurable sites of disease, were treated with concurrent radiotherapy (35 Gy in ten fractions, over 2 weeks) to one metastatic site and granulocyte-macrophage colony-stimulating factor (125 mug/m2 subcutaneously injected daily for 2 weeks, starting during the second week of radiotherapy). This course was repeated, targeting a second metastatic site. A Simon's optimal two-stage design was chosen for this trial: an additional 19 patients could be enrolled in stage 2 only if at least one patient among the first ten had an abscopal response. If no abscopal responses were seen among the first ten patients, the study would be deemed futile and terminated. The primary endpoint was the proportion of patients with an abscopal response (defined as at least a 30% decrease in the longest diameter of the best responding abscopal lesion). Secondary endpoints were safety and survival. Analyses were done based on intention to treat. The trial has concluded accrual, and is registered with ClinicalTrials.gov, number NCT02474186. FINDINGS: From April 7, 2003, to April 3, 2012, 41 patients with metastatic cancer were enrolled. In stage 1 of the Simon's two-stage design, ten patients were enrolled: four of the first ten patients had abscopal responses. Thus, the trial proceeded to stage 2, as planned, and an additional 19 patients were enrolled. Due to protocol amendments 12 further patients were enrolled. Abscopal responses occurred in eight (27.6%, 95% CI 12.7-47.2) of the first 29 patients, and 11 (26.8%, 95% CI 14.2-42.9) of 41 accrued patients (specifically in four patients with non-small-cell lung cancer, five with breast cancer, and two with thymic cancer). The most common grade 3-4 adverse events were fatigue (six patients) and haematological (ten patients). Additionally, a serious adverse event of grade 4 pulmonary embolism occurred in one patient. INTERPRETATION: The combination of radiotherapy with granulocyte-macrophage colony-stimulating factor produced objective abscopal responses in some patients with metastatic solid tumours. This finding represents a promising approach to establish an in-situ anti-tumour vaccine. Further research is warranted in this area. FUNDING: New York University School of Medicine's Department of Radiation Oncology and Cancer Institute.
PMID: 26095785
ISSN: 1474-5488
CID: 1640742
Raising the next generation of physician-scientists: the chairs' perspective [Letter]
Formenti, Silvia C; Bonner, James F; Hahn, Stephen M; Lawrence, Theodore S; Liu, Fei-Fei; Thomas, Charles R Jr
PMID: 25968820
ISSN: 1879-355x
CID: 1578782