12 results on '"Galon J"'
Search Results
2. Dissecting tumor lymphocyte infiltration to predict benefit from immune-checkpoint inhibitors in metastatic colorectal cancer: lessons from the AtezoT RIBE study.
- Author
-
Moretto R, Rossini D, Catteau A, Antoniotti C, Giordano M, Boccaccino A, Ugolini C, Proietti A, Conca V, Kassambara A, Pietrantonio F, Salvatore L, Lonardi S, Tamberi S, Tamburini E, Poma AM, Fieschi J, Fontanini G, Masi G, Galon J, and Cremolini C
- Subjects
- Humans, Immune Checkpoint Inhibitors pharmacology, Immune Checkpoint Inhibitors therapeutic use, Bevacizumab, Lymphocytes metabolism, Tumor Microenvironment, Colorectal Neoplasms, Colonic Neoplasms, Rectal Neoplasms
- Abstract
Background: Tumor immune cells influence the efficacy of immune-checkpoint inhibitors (ICIs) and many efforts aim at identifying features of tumor immune microenvironment able to predict benefit from ICIs in proficient mismatch repair (pMMR)/microsatellite stable (MSS) metastatic colorectal cancer (mCRC)., Methods: We characterized tumor immune cell infiltrate, by assessing tumor-infiltrating lymphocytes (TILs), Immunoscore, Immunoscore-IC, and programmed death ligand-1 (PD-L1) expression in tumor samples of patients with mCRC enrolled in the AtezoTRIBE study, a phase II randomized trial comparing FOLFOXIRI/bevacizumab/atezolizumab to FOLFOXIRI/bevacizumab, with the aim of evaluating the prognostic and predictive value of these features., Results: Out of 218 patients enrolled, 181 (83%), 77 (35%), 157 (72%) and 162 (74%) specimens were successfully tested for TILs, Immunoscore, Immunoscore-IC and PD-L1 expression, respectively, and 69 (38%), 45 (58%), 50 (32%) and 21 (13%) tumors were classified as TILs-high, Immunoscore-high, Immunoscore-IC-high and PD-L1-high, respectively. A poor agreement was observed between TILs and Immunoscore or Immunoscore-IC (K of Cohen <0.20). In the pMMR population, longer progression-free survival (PFS) was reported for Immunoscore-high and Immunoscore-IC-high groups compared with Immunoscore-low (16.4 vs 12.2 months; HR: 0.55, 95% CI: 0.30 to 0.99; p=0.049) and Immunoscore-IC-low (14.8 vs 11.5 months; HR: 0.55, 95% CI: 0.35 to 0.85; p=0.007), respectively, with a significant interaction effect between treatment arms and Immunoscore-IC (p for interaction: 0.006) and a trend for Immunoscore (p for interaction: 0.13). No PFS difference was shown according to TILs and PD-L1 expression. Consistent results were reported in the overall population., Conclusions: The digital evaluation of tumor immune cell infiltrate by means of Immunoscore-IC or Immunoscore identifies the subset of patients with pMMR mCRC achieving more benefit from the addition of the anti-PD-L1 to the upfront treatment. Immunoscore-IC stands as the most promising predictor of benefit from ICIs., Competing Interests: Competing interests: AC, AK, JF: are Veracyte employees. JG: has patents associated with the immune prognostic and predictive biomarkers, is co-founder of HalioDx, a Veracyte company. SL: has a consulting or an advisory role for Amgen, Merck Serono, Lilly, AstraZeneca, Incyte, Daiichi-Sankyo, BMS, Servier, and MSD; has received research funding from Amgen, Merck Serono, Bayer, Roche, Lilly, AstraZeneca, and BMS; and has received speakers’ fees from Roche, Lilly, BMS, Servier, Merck Serono, Pierre-Fabre, GlaxoSmithKline, and Amgen. FP: honoraria from Amgen, Bayer, Servier, Merck-Serono, Lilly, MSD, Organon, BMS, AstraZeneca, Pierre-Fabre; research grants from Bristol-Myers Squibb, AstraZeneca, Agenus and Incyte. LS: speakers’ and consultant’s fee from MSD, AstraZeneca, Servier, Bayer, Merck, Amgen, Pierre-Fabre. GM: received speakers’ fees—Merck, Amgen. CC: honoraria—Amgen, Bayer, Merck, Roche and Servier. Consulting or advisory role—Amgen, Bayer, MSD, Roche. Speakers’ Bureau—Servier. Research funding—Bayer, Merck, Servier. Travel, accommodations and expenses—Roche and Servier. All other authors have declared no conflicts of interest., (© Author(s) (or their employer(s)) 2023. Re-use permitted under CC BY-NC. No commercial re-use. See rights and permissions. Published by BMJ.)
- Published
- 2023
- Full Text
- View/download PDF
3. The Great Debate at 'Immunotherapy Bridge', Naples, December 5, 2019.
- Author
-
Ascierto PA, Bifulco C, Galon J, Garbe C, Khleif SN, McQuade J, Odunsi K, Okada H, Paulos CM, Quezada SA, Tawbi HA, Timmerman J, Trinchieri G, Butterfield LH, and Puzanov I
- Subjects
- History, 21st Century, Humans, Immunotherapy
- Abstract
As part of the 2019 Immunotherapy Bridge congress (December 4-5, Naples, Italy), the Great Debate session featured counterpoint views from leading experts on six topical issues in immunotherapy today. These were the use of chimeric antigen receptor T cell therapy in solid tumors, whether the Immunoscore should be more widely used in clinical practice, whether antibody-dependent cellular cytotoxicity is important in the mode of action of anticytotoxic T-lymphocyte-associated protein 4 antibodies, whether the brain is immunologically unique or just another organ, the role of microbiome versus nutrition in affecting responses to immunotherapy, and whether chemotherapy is immunostimulatory or immunosuppressive. Discussion of these important topics are summarized in this report., Competing Interests: Competing interests: PAA: consultant/advisory role for Bristol Myers-Squibb, Roche-Genentech, Merck Sharp & Dohme, Array, Novartis, Merck Serono, Pierre Fabre, Incyte, NewLink Genetics, Genmab, Medimmune, AstraZeneca, Syndax, SunPharma, Sanofi, Idera, Ultimovacs, Sandoz, Immunocore, 4SC, Alkermes, Italfarmaco, Nektar. He also received research funds from Bristol Myers-Squibb, Roche-Genentech, Array, and travel support from MSD. CB: scientific advisory/consulting for PrimeVax; research support: Heat Biologics. JG: co-founder and chairman of the scientific advisory board: HalioDx; collaborative Research Agreement (grants): Perkin-Elmer, IO Biotech, MedImmune, Janssen, Imcheck Therapeutics; participation to Scientific Advisory Boards: BMS, MedImmune, AstraZeneca, Novartis, Definiens, Merck Serono, IO Biotech, ImmunID, Nanostring, Illumina, Northwest Biotherapeutics, Actelion, Amgen, CatalYm, Merck MSD; Consultant: BMS, Roche, GSK, Compugen, Mologen, Gilead, Sanofi. CG: honoraria: Amgen, BMS, CeCaVa, MSD, NeraCare, Novartis, Philogen, Pierre Fabre, Roche, Sanofi; research grants: BMS, Novartis, Roche, Sanofi. SNK: advisory board/board member: Northwest Bio, UbiVac, Syndax Pharmaceuticals, PDS Biotechnology, KAHR, AratingaBio INC, IO Biotechnology, CanImGuide Therapeutics, Bioline Therapeutics, McKinsey Cancer Center, McKinsey and Company, Incyte, Cancer Panel, LLC Advaxis Immunetherapies; KOL/Consultant: J&J, AstraZeneca, NewLink Genetics, Medimmune, Lycera, Berringer Engelheim; unrestricted preclinical research funding: AstraZeneca, Bioline Therapeutics, Lycera, IO Biotech, Syndax, Biotechnologies. JMQ: funding: BMS, Merck. KO: in the past 2 years funding from: AstraZeneca, Tesaro Pharma; scientific advisory boards: Immunovaccine, Unleash Immuno-Oncolytics, Merck, Celsion, Truvax, Geneos, Triumvira; co-founder: Tactiva Therapeutics. HO: consultant for: Bristol-Myers Squibb, Alexion Pharmaceuticals, Gerson Lehrman Group, Amal Therapeutics, Agios Pharmaceuticals, Eureka Therapeutics, INC Research, LLC, LifeSci Capital, LLC; grant/research support from: Ono Pharmaceutical, Agios Pharmaceuticals and Midatech Pharma; inventor of: the H3.3K27M TCR, IL-13Ra2 (345-353:1A9V) peptide, EGFRvIII-CAR as well as Rheo-IL-12 for which an exclusive licensing agreement has been executed with Tmunity, Inc., Stemline, Inc., Novartis Pharma and Intrexon Corporation, respectively. CMP: Lycera, Ares Immunotherapy, Thermo Fisher, Obsidian Therapeutics. SAQ: funder of Achilles therapeutics; research funding: Tusk Therapeutics; scientific advisor: Achilles therapeutics, Tusk Therapeutics/Roche, Morphosys, Bicycle Therapeutics, Ioncturas. HAT: research funding and consulting fees listed below from the following sources: BMS, Novartis, Genentech/Roche, Merck, Array, Celgene, GSK. JT: research support from Kite/Gilead, Merck, BMS, Spectrum Pharmaceuticals; advisory board member/consultant for Kite/Gilead, BMS, Celgene. GT: declares he has not received funding or has relationships with subjects with commercial interests in the health field. LHB: scientific and medical advisory board: StemImmune/Calidi, SapVax, NextCure, Replimmune, Western Oncolytics, Torque Therapeutics, Khloris, Pyxis, Cytomix. IP: consultant, research funding to institutional Amgen, Nektar, ADC, Idera Pharmaceutical., (© Author(s) (or their employer(s)) 2020. Re-use permitted under CC BY-NC. No commercial re-use. See rights and permissions. Published by BMJ.)
- Published
- 2020
- Full Text
- View/download PDF
4. Analytical validation of the Immunoscore and its associated prognostic value in patients with colon cancer.
- Author
-
Marliot F, Chen X, Kirilovsky A, Sbarrato T, El Sissy C, Batista L, Van den Eynde M, Haicheur-Adjouri N, Anitei MG, Musina AM, Scripcariu V, Lagorce-Pagès C, Hermitte F, Galon J, Fieschi J, and Pagès F
- Subjects
- Female, Humans, Male, Prognosis, Reproducibility of Results, Colonic Neoplasms immunology
- Abstract
Background: New and fully validated tests need to be brought into clinical practice to improve the estimation of recurrence risk in patients with colon cancer. The aim of this study was to assess the analytical performances of the Immunoscore (IS) and show its contribution to prognosis prediction., Methods: Immunohistochemical staining of CD3+ and CD8+ T cells on adjacent sections of colon cancer tissues were quantified in the core of the tumor and its invasive margin with dedicated IS modules integrated into digital pathology software. Staining intensity across samples collected between 1989 and 2016 (n=595) was measured. The accuracy of the IS workflow was established by comparing optical and automatic counts. Analytical precision of the IS was evaluated within individual tumor block on distant sections and between eligible blocks. The IS interlaboratory reproducibility (n=100) and overall assay precision were assessed (n=3). Contribution of the IS to prediction of recurrence based on clinical and molecular parameters was determined (n=538)., Results: Optical and automatic counts for CD3+ or CD8+ were strongly correlated (r=0.94, p<0.001 and r=0.92, p<0.001, respectively). CD3 and CD8 staining intensities were not altered by the age of the tumor block over a period of 30 years. Neither the position of tested tissue sections within a tumor block nor the selection of the tissue blocks affected the IS. Reproducibility of the IS was not affected by multiple variables (eg, antibody lots, DAB revelation kits, immunohistochemistry automates and operators). Interassay repeatability of the IS was 100% and interlaboratory reproducibility between two testing centers was 93%. Finally, in a case series of patients with stage II-III colon cancer, the relative proportion of variance for time to recurrence was greatest for the IS (53% of prognostic variability) in a model that included IS, T-stage, microsatellite instability status and total number of lymph nodes., Conclusion: IS is a robust and validated clinical assay leveraging immune scoring to predict recurrence risk of patient with localized colon cancer. The strong and independent prognostic value of IS should pave the way for it use in clinical practice., Competing Interests: Competing interests: JG and FP have patents associated with the immune prognostic biomarkers. JG and FH are HalioDx co-founders and shareholders. All other authors declare no competing interests. The IS is a registered trademark owned by the National Institute of Health and Medical Research., (© Author(s) (or their employer(s)) 2020. Re-use permitted under CC BY-NC. No commercial re-use. See rights and permissions. Published by BMJ.)
- Published
- 2020
- Full Text
- View/download PDF
5. Oncogenic states dictate the prognostic and predictive connotations of intratumoral immune response.
- Author
-
Roelands J, Hendrickx W, Zoppoli G, Mall R, Saad M, Halliwill K, Curigliano G, Rinchai D, Decock J, Delogu LG, Turan T, Samayoa J, Chouchane L, Ballestrero A, Wang E, Finetti P, Bertucci F, Miller LD, Galon J, Marincola FM, Kuppen PJK, Ceccarelli M, and Bedognetti D
- Subjects
- Female, Humans, Male, Neoplasms mortality, Prognosis, Survival Analysis, Gene Expression Profiling methods, Immunity immunology, Immunotherapy methods, Neoplasms immunology, Oncogenes immunology
- Abstract
Background: An immune active cancer phenotype typified by a T helper 1 (Th-1) immune response has been associated with increased responsiveness to immunotherapy and favorable prognosis in some but not all cancer types. The reason of this differential prognostic connotation remains unknown., Methods: To explore the contextual prognostic value of cancer immune phenotypes, we applied a multimodal pan-cancer analysis among 31 different histologies (9282 patients), encompassing immune and oncogenic transcriptomic analysis, mutational and neoantigen load and copy number variations., Results: We demonstrated that the favorable prognostic connotation conferred by the presence of a Th-1 immune response was abolished in tumors displaying specific tumor-cell intrinsic attributes such as high transforming growth factor-beta (TGF-β) signaling and low proliferation capacity. This observation was independent of mutation rate. We validated this observation in the context of immune checkpoint inhibition. WNT- β catenin, barrier molecules, Notch, hedgehog, mismatch repair, telomerase activity and AMPK signaling were the pathways most coherently associated with an immune silent phenotype together with mutations of driver genes including IDH1/2, FOXA2, HDAC3, PSIP1, MAP3K1, KRAS, NRAS, EGFR, FGFR3, WNT5A and IRF7 ., Conclusions: This is the first systematic study demonstrating that the prognostic and predictive role of a bona fide favorable intratumoral immune response is dependent on the disposition of specific oncogenic pathways. This information could be used to refine stratification algorithms and prioritize hierarchically relevant targets for combination therapies., Competing Interests: Competing interests: TT, KH, and JS are employees of AbbVie Biotherapeutics. MC was an employee of AbbVie Biotherapeutics at the time the research was conducted and has currently a consulting relationship with AbbVie. FMM is an employee of Refuge Biotechnologies. EW is an employee of Allogene Therapeutics, (© Author(s) (or their employer(s)) 2020. Re-use permitted under CC BY-NC. No commercial re-use. See rights and permissions. Published by BMJ.)
- Published
- 2020
- Full Text
- View/download PDF
6. Perspectives in immunotherapy: meeting report from the "Immunotherapy Bridge 2018" (28-29 November, 2018, Naples, Italy).
- Author
-
Ascierto PA, Bifulco C, Buonaguro L, Emens LA, Ferris RL, Fox BA, Delgoffe GM, Galon J, Gridelli C, Merlano M, Nathan P, Odunsi K, Okada H, Paulos CM, Pignata S, Schalper KA, Spranger S, Tortora G, Zarour H, Butterfield LH, and Puzanov I
- Subjects
- Humans, Molecular Targeted Therapy, Neoplasms etiology, Neoplasms metabolism, T-Lymphocytes drug effects, Antineoplastic Agents, Immunological pharmacology, Antineoplastic Agents, Immunological therapeutic use, Biomarkers, Tumor, Immunotherapy methods, Neoplasms immunology, Neoplasms therapy, T-Lymphocytes immunology, T-Lymphocytes metabolism
- Abstract
Immunotherapy is now widely established as a potent and effective treatment option across several types of cancer. However, there is increasing recognition that not all patients respond to immunotherapy, focusing attention on the immune contexture of the tumor microenvironment (TME), drivers of the immune response and mechanisms of tumor resistance to immunity. The development of novel immunotherapeutics and their use in combination with checkpoint inhibitors and other standard of care and novel treatment modalities is an area of particular attention across several tumor types, including melanoma, lung, ovarian, breast, pancreatic, renal, head and neck, brain and non-melanoma skin cancers. The 4th Immunotherapy Bridge meeting (28-29 November, 2018, Naples, Italy) focused on a wide range of evolving topics and trends in the field of cancer immunotherapy and key presentations from this meeting are summarised in this report.
- Published
- 2019
- Full Text
- View/download PDF
7. Correction to: Toward a comprehensive view of cancer immune responsiveness: a synopsis from the SITC workshop.
- Author
-
Bedognetti D, Ceccarelli M, Galluzzi L, Lu R, Palucka K, Samayoa J, Spranger S, Warren S, Wong KK, Ziv E, Chowell D, Coussens LM, De Carvalho DD, DeNardo DG, Galon J, Kaufman HL, Kirchhoff T, Lotze MT, Luke JJ, Minn AJ, Politi K, Shultz LD, Simon R, Thórsson V, Weidhaas JB, Ascierto ML, Ascierto PA, Barnes JM, Barsan V, Bommareddy PK, Bot A, Church SE, Ciliberto G, De Maria A, Draganov D, Ho WS, McGee HM, Monette A, Murphy JF, Nisticò P, Park W, Patel M, Quigley M, Radvanyi L, Raftopoulos H, Rudqvist NP, Snyder A, Sweis RF, Valpione S, Zappasodi R, Butterfield LH, Disis ML, Fox BA, Cesano A, and Marincola FM
- Abstract
Following publication of the original article [1], the author reported that an author name, Roberta Zappasodi, was missed in the authorship list.
- Published
- 2019
- Full Text
- View/download PDF
8. Toward a comprehensive view of cancer immune responsiveness: a synopsis from the SITC workshop.
- Author
-
Bedognetti D, Ceccarelli M, Galluzzi L, Lu R, Palucka K, Samayoa J, Spranger S, Warren S, Wong KK, Ziv E, Chowell D, Coussens LM, De Carvalho DD, DeNardo DG, Galon J, Kaufman HL, Kirchhoff T, Lotze MT, Luke JJ, Minn AJ, Politi K, Shultz LD, Simon R, Thórsson V, Weidhaas JB, Ascierto ML, Ascierto PA, Barnes JM, Barsan V, Bommareddy PK, Bot A, Church SE, Ciliberto G, De Maria A, Draganov D, Ho WS, McGee HM, Monette A, Murphy JF, Nisticò P, Park W, Patel M, Quigley M, Radvanyi L, Raftopoulos H, Rudqvist NP, Snyder A, Sweis RF, Valpione S, Zappasodi R, Butterfield LH, Disis ML, Fox BA, Cesano A, and Marincola FM
- Subjects
- Advisory Committees, Animals, Biomarkers, Tumor genetics, Biomarkers, Tumor immunology, Congresses as Topic, Disease Models, Animal, Humans, Medical Oncology organization & administration, Neoplasms genetics, Neoplasms immunology, Societies, Medical organization & administration, Treatment Outcome, Tumor Microenvironment genetics, Immunotherapy, Neoplasms therapy, Tumor Microenvironment immunology
- Abstract
Tumor immunology has changed the landscape of cancer treatment. Yet, not all patients benefit as cancer immune responsiveness (CIR) remains a limitation in a considerable proportion of cases. The multifactorial determinants of CIR include the genetic makeup of the patient, the genomic instability central to cancer development, the evolutionary emergence of cancer phenotypes under the influence of immune editing, and external modifiers such as demographics, environment, treatment potency, co-morbidities and cancer-independent alterations including immune homeostasis and polymorphisms in the major and minor histocompatibility molecules, cytokines, and chemokines. Based on the premise that cancer is fundamentally a disorder of the genes arising within a cell biologic process, whose deviations from normality determine the rules of engagement with the host's response, the Society for Immunotherapy of Cancer (SITC) convened a task force of experts from various disciplines including, immunology, oncology, biophysics, structural biology, molecular and cellular biology, genetics, and bioinformatics to address the complexity of CIR from a holistic view. The task force was launched by a workshop held in San Francisco on May 14-15, 2018 aimed at two preeminent goals: 1) to identify the fundamental questions related to CIR and 2) to create an interactive community of experts that could guide scientific and research priorities by forming a logical progression supported by multiple perspectives to uncover mechanisms of CIR. This workshop was a first step toward a second meeting where the focus would be to address the actionability of some of the questions identified by working groups. In this event, five working groups aimed at defining a path to test hypotheses according to their relevance to human cancer and identifying experimental models closest to human biology, which include: 1) Germline-Genetic, 2) Somatic-Genetic and 3) Genomic-Transcriptional contributions to CIR, 4) Determinant(s) of Immunogenic Cell Death that modulate CIR, and 5) Experimental Models that best represent CIR and its conversion to an immune responsive state. This manuscript summarizes the contributions from each group and should be considered as a first milestone in the path toward a more contemporary understanding of CIR. We appreciate that this effort is far from comprehensive and that other relevant aspects related to CIR such as the microbiome, the individual's recombined T cell and B cell receptors, and the metabolic status of cancer and immune cells were not fully included. These and other important factors will be included in future activities of the taskforce. The taskforce will focus on prioritization and specific actionable approach to answer the identified questions and implementing the collaborations in the follow-up workshop, which will be held in Houston on September 4-5, 2019.
- Published
- 2019
- Full Text
- View/download PDF
9. Perspectives in immunotherapy: meeting report from the Immunotherapy Bridge (29-30 November, 2017, Naples, Italy).
- Author
-
Ascierto PA, Brugarolas J, Buonaguro L, Butterfield LH, Carbone D, Daniele B, Ferris R, Fox BA, Galon J, Gridelli C, Kaufman HL, Klebanoff CA, Melero I, Nathan P, Paulos CM, Ruella M, Sullivan R, Zarour H, and Puzanov I
- Subjects
- Animals, Antigen Presentation immunology, Antineoplastic Agents, Immunological pharmacology, Antineoplastic Agents, Immunological therapeutic use, Biomarkers, Tumor, CTLA-4 Antigen antagonists & inhibitors, Cancer Vaccines immunology, Humans, Immunomodulation drug effects, Liquid Biopsy, Mutation, Neoplasms diagnosis, Neoplasms metabolism, Programmed Cell Death 1 Receptor antagonists & inhibitors, T-Lymphocytes drug effects, T-Lymphocytes immunology, T-Lymphocytes metabolism, Translational Research, Biomedical, Tumor Microenvironment drug effects, Tumor Microenvironment genetics, Tumor Microenvironment immunology, Immunotherapy methods, Neoplasms immunology, Neoplasms therapy
- Abstract
Immunotherapy represents the third important wave in the history of the systemic treatment of cancer after chemotherapy and targeted therapy and is now established as a potent and effective treatment option across several cancer types. The clinical success of anti-cytotoxic T-lymphocyte-associated antigen (CTLA)-4, first, and anti-programmed death (PD)-1/PD-ligand (L)1 agents in melanoma and other cancers a few years later, has encouraged increasing focus on the development of other immunotherapies (e.g. monoclonal antibodies with other immune targets, adoptive cell transfer, and vaccines), with over 3000 immuno-oncology trials ongoing, involving hundreds of research institutes across the globe. The potential use of these different immunotherapeutic options in various combinations with one another and with other treatment modalities is an area of particular promise. The third Immunotherapy Bridge meeting (29-30 November, 2017, Naples, Italy) focused on recent advances in immunotherapy across various cancer types and is summarised in this report.
- Published
- 2018
- Full Text
- View/download PDF
10. Identifying baseline immune-related biomarkers to predict clinical outcome of immunotherapy.
- Author
-
Gnjatic S, Bronte V, Brunet LR, Butler MO, Disis ML, Galon J, Hakansson LG, Hanks BA, Karanikas V, Khleif SN, Kirkwood JM, Miller LD, Schendel DJ, Tanneau I, Wigginton JM, and Butterfield LH
- Subjects
- Antigens, Neoplasm analysis, B-Lymphocytes immunology, Humans, Mutation, Myeloid-Derived Suppressor Cells immunology, Neoplasms genetics, Neoplasms immunology, Prognosis, Tumor Microenvironment immunology, Biomarkers, Tumor analysis, Immunotherapy methods, Neoplasms therapy
- Abstract
As cancer strikes, individuals vary not only in terms of factors that contribute to its occurrence and development, but as importantly, in their capacity to respond to treatment. While exciting new therapeutic options that mobilize the immune system against cancer have led to breakthroughs for a variety of malignancies, success is limited to a subset of patients. Pre-existing immunological features of both the host and the tumor may contribute to how patients will eventually fare with immunotherapy. A broad understanding of baseline immunity, both in the periphery and in the tumor microenvironment, is needed in order to fully realize the potential of cancer immunotherapy. Such interrogation of the tumor, blood, and host immune parameters prior to treatment is expected to identify biomarkers predictive of clinical outcome as well as to elucidate why some patients fail to respond to immunotherapy. To approach these opportunities for progress, the Society for Immunotherapy of Cancer (SITC) reconvened the Immune Biomarkers Task Force. Comprised of an international multidisciplinary panel of experts, Working Group 4 sought to make recommendations that focus on the complexity of the tumor microenvironment, with its diversity of immune genes, proteins, cells, and pathways naturally present at baseline and in circulation, and novel tools to aid in such broad analyses.
- Published
- 2017
- Full Text
- View/download PDF
11. Society for immunotherapy of cancer (SITC) statement on the proposed changes to the common rule.
- Author
-
Kaufman HL, Butterfield LH, Coulie PG, Demaria S, Ferris RL, Galon J, Khleif SN, Mellman I, Ohashi PS, Overwijk WW, Topalian SL, and Marincola FM
- Abstract
The Common Rule is a set of ethical principles that provide guidance on the management of human subjects taking part in biomedical and behavioral research in the United States. The elements of the Common Rule were initially developed in 1981 following a revision of the Declaration of Helsinki in 1975. Most academic facilities follow the Common Rule in the regulation of clinical trials research. Recently, the government has suggested a revision of the Common Rule to include more contemporary and streamlined oversight of clinical research. In this commentary, the leadership of the Society for Immunotherapy of Cancer (SITC) provides their opinion on this plan. While the Society recognizes the considerable contribution of clinical research in supporting progress in tumor immunotherapy and supports the need for revisions to the Common Rule, there is also some concern over certain elements which may restrict access to biospecimens and clinical data at a time when high throughput technologies, computational biology and assay standardization is allowing major advances in understanding cancer biology and providing potential predictive biomarkers of immunotherapy response. The Society values its professional commitment to patients for improving clinical outcomes with tumor immunotherapy and supports continued discussion with all stakeholders before implementing changes to the Common Rule in order to ensure maximal patient protections while promoting continued clinical research at this historic time in cancer research.
- Published
- 2016
- Full Text
- View/download PDF
12. Immunodynamics: a cancer immunotherapy trials network review of immune monitoring in immuno-oncology clinical trials.
- Author
-
Kohrt HE, Tumeh PC, Benson D, Bhardwaj N, Brody J, Formenti S, Fox BA, Galon J, June CH, Kalos M, Kirsch I, Kleen T, Kroemer G, Lanier L, Levy R, Lyerly HK, Maecker H, Marabelle A, Melenhorst J, Miller J, Melero I, Odunsi K, Palucka K, Peoples G, Ribas A, Robins H, Robinson W, Serafini T, Sondel P, Vivier E, Weber J, Wolchok J, Zitvogel L, Disis ML, and Cheever MA
- Abstract
The efficacy of PD-1/PD-L1 targeted therapies in addition to anti-CTLA-4 solidifies immunotherapy as a modality to add to the anticancer arsenal. Despite raising the bar of clinical efficacy, immunologically targeted agents raise new challenges to conventional drug development paradigms by highlighting the limited relevance of assessing standard pharmacokinetics (PK) and pharmacodynamics (PD). Specifically, systemic and intratumoral immune effects have not consistently correlated with standard relationships between systemic dose, toxicity, and efficacy for cytotoxic therapies. Hence, PK and PD paradigms remain inadequate to guide the selection of doses and schedules, both starting and recommended Phase 2 for immunotherapies. The promise of harnessing the immune response against cancer must also be considered in light of unique and potentially serious toxicities. Refining immune endpoints to better inform clinical trial design represents a high priority challenge. The Cancer Immunotherapy Trials Network investigators review the immunodynamic effects of specific classes of immunotherapeutic agents to focus immune assessment modalities and sites, both systemic and importantly intratumoral, which are critical to the success of the rapidly growing field of immuno-oncology.
- Published
- 2016
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.