Melanoma and immunotherapy bridge 2015 : Naples, Italy. 1-5 December 2015
1,3,5,6,11,12,14,18,19,21,22,23,27,29,30,31,36,37,38,39,40,42,43,44,46,49,52,55,56,57,58,64,65,66,68,69,72,73,74,75,76,77,78,79,80,82,83,85,86,87,91,93,96,97,98,99,100,101,104,106,107,108,109,110,111,2,70,112,113,114,115,116,16,117,119,120,121,123,124,125,126,127,128,129,118,130,131,132,133,134,135,137,138,139,140,141,142,143,144,145,146,147,148,149,150,151,152,153,154,156,158,160,162,167,170,171,172,173,176,177,185,186,88,187,188,61,189,184,190,192,193,196,197,90,9,202,206,92,207,208,209,203,210,13,211,212,213,214,215,4,216,217,157,159,218,219,220,221,222,223,224,53,225,54,204,226,227,10,51,228,229,230,231,232,233,234,235,236,237,239,240,242,243,244,245,246,248
DOI: https://doi.org/10.1186/s12967-016-0791-2
2016-07-25
Abstract:MELANOMA BRIDGE 2015 KEYNOTE SPEAKER PRESENTATIONS Molecular and immuno-advances K1 Immunologic and metabolic consequences of PI3K/AKT/mTOR activation in melanoma Vashisht G. Y. Nanda, Weiyi Peng, Patrick Hwu, Michael A. Davies K2 Non-mutational adaptive changes in melanoma cells exposed to BRAF and MEK inhibitors help the establishment of drug resistance Gennaro Ciliberto, Luigi Fattore, Debora Malpicci, Luigi Aurisicchio, Paolo Antonio Ascierto, Carlo M. Croce, Rita Mancini K3 Tumor-intrinsic beta-catenin signaling mediates tumor-immune avoidance Stefani Spranger, Thomas F. Gajewski K4 Intracellular tumor antigens as a source of targets of antibody-based immunotherapy of melanoma Yangyang Wang, Soldano Ferrone Combination therapies K5 Harnessing radiotherapy to improve responses to immunotherapy in cancer Claire Vanpouille-Box, Erik Wennerberg, Karsten A. Pilones, Silvia C. Formenti, Sandra Demaria K6 Creating a T cell-inflamed tumor microenvironment overcomes resistance to checkpoint blockade Haidong Tang, Yang Wang, Yang-Xin Fu K7 Biomarkers for treatment decisions? Reinhard Dummer K8 Combining oncolytic therapies in the era of checkpoint inhibitors Igor Puzanov K9 Immune checkpoint blockade for melanoma: should we combine or sequence ipilimumab and PD-1 antibody therapy? Michael A. Postow News in immunotherapy K10 An update on adjuvant and neoadjuvant therapy for melanom Ahmad Tarhini K11 Targeting multiple inhibitory receptors in melanoma Joe-Marc Chauvin, Ornella Pagliano, Julien Fourcade, Zhaojun Sun, Hong Wang, Cindy Sanders, John M. Kirkwood, Tseng-hui Timothy Chen, Mark Maurer, Alan J. Korman, Hassane M. Zarour K12 Improving adoptive immune therapy using genetically engineered T cells David F. Stroncek Tumor microenvironment and biomarkers K13 Myeloid cells and tumor exosomes: a crosstalk for assessing immunosuppression? Veronica Huber, Licia Rivoltini K14 Update on the SITC biomarker taskforce: progress and challenges Magdalena Thurin World-wide immunoscore task force: an update K15 The immunoscore in colorectal cancer highlights the importance of digital scoring systems in surgical pathology Tilman Rau, Alessandro Lugli K16 The immunoscore: toward an integrated immunomonitoring from the diagnosis to the follow up of cancer’s patients Franck Pagès Economic sustainability of melanoma treatments: regulatory, health technology assessment and market access issues K17 Nivolumab, the regulatory experience in immunotherapy Jorge Camarero, Arantxa Sancho K18 Evidence to optimize access for immunotherapies Claudio Jommi ORAL PRESENTATIONS Molecular and immuno-advances O1 Ipilimumab treatment results in CD4 T cell activation that is concomitant with a reduction in Tregs and MDSCs Yago Pico de Coaña, Maria Wolodarski, Yuya Yoshimoto, Giusy Gentilcore, Isabel Poschke, Giuseppe V. Masucci, Johan Hansson, Rolf Kiessling O2 Evaluation of prognostic and therapeutic potential of COX-2 and PD-L1 in primary and metastatic melanoma Giosuè Scognamiglio, Francesco Sabbatino, Federica Zito Marino, Anna Maria Anniciello, Monica Cantile, Margherita Cerrone, Stefania Scala, Crescenzo D’alterio, Angela Ianaro, Giuseppe Cirino, Paolo Antonio Ascierto, Giuseppina Liguori, Gerardo Botti O3 Vemurafenib in patients with BRAFV600 mutation–positive metastatic melanoma: final overall survival results of the BRIM-3 study Paul B. Chapman, Caroline Robert, James Larkin, John B. Haanen, Antoni Ribas, David Hogg, Omid Hamid, Paolo Antonio Ascierto, Alessandro Testori, Paul Lorigan, Reinhard Dummer, Jeffrey A. Sosman, Keith T. Flaherty, Huibin Yue, Shelley Coleman, Ivor Caro, Axel Hauschild, Grant A. McArthur O4 Updated survival, response and safety data in a phase 1 dose-finding study (CA209-004) of concurrent nivolumab (NIVO) and ipilimumab (IPI) in advanced melanoma Mario Sznol, Margaret K. Callahan, Harriet Kluger, Michael A. Postow, RuthAnn Gordan, Neil H. Segal, Naiyer A. Rizvi, Alexander Lesokhin, Michael B. Atkins, John M. Kirkwood, Matthew M. Burke, Amanda Ralabate, Angel Rivera, Stephanie A. Kronenberg, Blessing Agunwamba, Mary Ruisi, Christine Horak, Joel Jiang, Jedd Wolchok Combination therapies O5 Efficacy and correlative biomarker analysis of the coBRIM study comparing cobimetinib (COBI) + vemurafenib (VEM) vs placebo (PBO) + VEM in advanced BRAF-mutated melanoma patients (pts) Paolo A. Ascierto, Grant A. McArthur, James Larkin, Gabriella Liszkay, Michele Maio, Mario Mandalà, Lev Demidov, Daniil Stoyakovskiy, Luc Thomas, Luis de la Cruz-Merino, Victoria Atkinson, Caroline Dutriaux, Claus Garbe, Matthew Wongchenko, Ilsung Chang, Daniel O. Koralek, Isabelle Rooney, Yibing Yan, Antoni Ribas, Brigitte Dréno O6 Preliminary clinical safety, tolerability and activity results from a Phase Ib study of atezolizumab (anti-PDL1) combined with vemurafenib in BRAFV600-mutant metastatic melanoma Ryan Sullivan, Omid Hamid, Manish Patel, Stephen Hodi, Rodabe Amaria, Peter Boasberg, Jeffrey Wallin, Xian He, Edward Cha, Nicole Richie, Marcus Ballinger, Patrick Hwu O7 Preliminary safety and efficacy data from a phase 1/2 study of epacadostat (INCB024360) in combination with pembrolizumab in patients with advanced/metastatic melanoma Thomas F. Gajewski, Omid Hamid, David C. Smith, Todd M. Bauer, Jeffrey S. Wasser, Jason J. Luke, Ani S. Balmanoukian, David R. Kaufman, Yufan Zhao, Janet Maleski, Lance Leopold, Tara C. Gangadhar O8 Primary analysis of MASTERKEY-265 phase 1b study of talimogene laherparepvec (T-VEC) and pembrolizumab (pembro) for unresectable stage IIIB-IV melanoma Reinhard Dummer, Georgina V. Long, Antoni Ribas, Igor Puzanov, Olivier Michielin, Ari VanderWalde, Robert H.I. Andtbacka, Jonathan Cebon, Eugenio Fernandez, Josep Malvehy, Anthony J. Olszanski, Thomas F. Gajewski, John M. Kirkwood, Christine Gause, Lisa Chen, David R. Kaufman, Jeffrey Chou, F. Stephen Hodi News in immunotherapy O9 Two-year survival and safety update in patients (pts) with treatment-naïve advanced melanoma (MEL) receiving nivolumab (NIVO) or dacarbazine (DTIC) in CheckMate 066 Victoria Atkinson, Paolo A. Ascierto, Georgina V. Long, Benjamin Brady, Caroline Dutriaux, Michele Maio, Laurent Mortier, Jessica C. Hassel, Piotr Rutkowski, Catriona McNeil, Ewa Kalinka-Warzocha, Celeste Lebbé, Lars Ny, Matias Chacon, Paola Queirolo, Carmen Loquai, Parneet Cheema, Alfonso Berrocal, Karmele Mujika Eizmendi, Luis De La Cruz-Merino, Gil Bar-Sela, Christine Horak, Joel Jiang, Helene Hardy, Caroline Robert O10 Efficacy and safety of nivolumab (NIVO) in patients (pts) with advanced melanoma (MEL) who were treated beyond progression in CheckMate 066/067 Georgina V. Long, Jeffrey S. Weber, James Larkin, Victoria Atkinson, Jean-Jacques Grob, Reinhard Dummer, Caroline Robert, Ivan Marquez-Rodas, Catriona McNeil, Henrik Schmidt, Karen Briscoe, Jean-François Baurain, F. Stephen Hodi, Jedd D. Wolchok Tumor microenvironment and biomarkers O11 New biomarkers for response/resistance to BRAF inhibitor therapy in metastatic melanoma Rosamaria Pinto, Simona De Summa, Vito Michele Garrisi, Sabino Strippoli, Amalia Azzariti, Gabriella Guida, Michele Guida, Stefania Tommasi O12 Chemokine receptor patterns in lymphocytes mirror metastatic spreading in melanoma and response to ipilimumab Nicolas Jacquelot, David Enot, Caroline Flament, Jonathan M. Pitt, Nadège Vimond, Carolin Blattner, Takahiro Yamazaki, Maria-Paula Roberti, Marie Vetizou, Romain Daillere, Vichnou Poirier-Colame, Michaëla Semeraro, Anne Caignard, Craig L Slingluff Jr, Federica Sallusto, Sylvie Rusakiewicz, Benjamin Weide, Aurélien Marabelle, Holbrook Kohrt, Stéphane Dalle, Andréa Cavalcanti, Guido Kroemer, Anna Maria Di Giacomo, Michaele Maio, Phillip Wong, Jianda Yuan, Jedd Wolchok, Viktor Umansky, Alexander Eggermont, Laurence Zitvogel O13 Serum levels of PD1- and CD28-positive exosomes before Ipilimumab correlate with therapeutic response in metastatic melanoma patients Passarelli Anna, Tucci Marco, Stucci Stefania, Mannavola Francesco, Capone Mariaelena, Madonna Gabriele, Ascierto Paolo Antonio, Silvestris Franco O14 Immunological prognostic factors in stage III melanomas María Paula Roberti, Nicolas Jacquelot, David P Enot, Sylvie Rusakiewicz, Michaela Semeraro, Sarah Jégou, Camila Flores, Lieping Chen, Byoung S. Kwon, Ana Carrizossa Anderson, Caroline Robert, Christophe Borg, Benjamin Weide, François Aubin, Stéphane Dalle, Michele Maio, Jedd D. Wolchok, Holbrook Kohrt, Maha Ayyoub, Guido Kroemer, Aurélien Marabelle, Andréa Cavalcanti, Alexander Eggermont, Laurence Zitvogel POSTER PRESENTATIONS Molecular and immuno-advances P1 Human melanoma cells resistant to B-RAF and MEK inhibition exhibit mesenchymal-like features Anna Lisa De Presbiteris, Fabiola Gilda Cordaro, Rosa Camerlingo, Federica Fratangelo, Nicola Mozzillo, Giuseppe Pirozzi, Eduardo J. Patriarca, Paolo A. Ascierto, Emilia Caputo P2 Anti-proliferative and pro-apoptotic effect of ABT888 on melanoma cell lines and its potential role in the treatment of melanoma resistant to B-RAF inhibitors Federica Fratangelo, Rosa Camerlingo, Emilia Caputo, Maria Letizia Motti, Rosaria Falcone, Roberta Miceli, Mariaelena Capone, Gabriele Madonna, Domenico Mallardo, Maria Vincenza Carriero, Giuseppe Pirozzi and Paolo Antonio Ascierto P3 Involvement of the L-cysteine/CSE/H2S pathway in human melanoma progression Elisabetta Panza, Paola De Cicco, Chiara Armogida, Giuseppe Ercolano, Rosa Camerlingo, Giuseppe Pirozzi, Giosuè Scognamiglio, Gerardo Botti, Giuseppe Cirino, Angela Ianaro P4 Cancer stem cell antigen revealing pattern of antibody variable region genes were defined by immunoglobulin repertoire analysis in patients with malignant melanoma Beatrix Kotlan, Gabriella Liszkay, Miri Blank, Timea Balatoni, Judit Olasz, Emil Farkas, Andras Szollar, Akos Savolt, Maria Godeny, Orsolya Csuka, Szabolcs Horvath, Klara Eles, Yehuda Shoenfeld and Miklos Kasler P5 Upregulation of Neuregulin-1 expression is a hallmark of adaptive response to BRAF/MEK inhibitors in melanoma Debora Malpicci, Luigi Fattore, Susan Costantini, Francesca Capone, Paolo Antonio Ascierto, Rita Mancini, Gennaro Ciliberto P6 HuR positively regulates migration of HTB63 melanoma cells Farnaz Moradi, Pontus Berglund, Karin Leandersson, Rickard Linnskog, Tommy Andersson, Chandra Prakash Prasad P7 Prolyl 4- (C-P4H) hydroxylases have opposing effects in malignant melanoma: implication in prognosis and therapy Cristiana Lo Nigro, Laura Lattanzio, Hexiao Wang, Charlotte Proby, Nelofer Syed, Marcella Occelli, Carolina Cauchi, Marco Merlano, Catherine Harwood, Alastair Thompson, Tim Crook P8 Urokinase receptor antagonists: novel agents for the treatment of melanoma Maria Letizia Motti, Katia Bifulco, Vincenzo Ingangi, Michele Minopoli, Concetta Ragone, Federica Fratangelo, Antonello Pessi, Gennaro Ciliberto, Paolo Antonio Ascierto, Maria Vincenza Carriero P9 Exosomes released by melanoma cell lines enhance chemotaxis of primary tumor cells Francesco Mannavola, Stella D’Oronzo, Claudia Felici, Marco Tucci, Antonio Doronzo, Franco Silvestris P10 New insights in mitochondrial metabolic reprogramming in melanoma Anna Ferretta, Gabriella Guida, Stefania Guida, Imma Maida, Tiziana Cocco, Sabino Strippoli, Stefania Tommasi, Amalia Azzariti, Michele Guida P11 Lenalidomide restrains the proliferation in melanoma cells through a negative regulation of their cell cycle Stella D’Oronzo, Anna Passarelli, Claudia Felici, Marco Tucci, Davide Quaresmini, Franco Silvestris Combination therapies P12 Chemoimmunotherapy elicits polyfunctional anti-tumor CD8 + T cells depending on the activation of an AKT pathway sustained by ICOS Ornella Franzese, Belinda Palermo, Cosmo Di Donna, Isabella Sperduti, MariaLaura Foddai, Helena Stabile, Angela Gismondi, Angela Santoni, Paola Nisticò P13 Favourable toxicity profile of combined BRAF and MEK inhibitors in metastatic melanoma patients Andrea P. Sponghini, Francesca Platini, Elena Marra, David Rondonotti, Oscar Alabiso, Maria T. Fierro, Paola Savoia, Florian Stratica, Pietro Quaglino P14 Electrothermal bipolar vessel sealing system dissection reduces seroma output or time to drain removal following axillary and ilio-inguinal node dissection in melanoma patients: a pilot study Di Monta Gianluca, Caracò Corrado, Di Marzo Massimiliano, Marone Ugo, Di Cecilia Maria Luisa, Mozzillo Nicola News in immunotherapy P15 Clinical and immunological response to ipilimumab in a metastatic melanoma patient with HIV infection Francesco Sabbatino, Celeste Fusciello1, Antonio Marra, Rosario Guarrasi, Carlo Baldi, Rosa Russo, Di Giulio Giovanni, Vincenzo Faiola, Pio Zeppa, Stefano Pepe P16 Immunotherapy and hypophysitis: a case report Elisabetta Gambale, Consiglia Carella, Alessandra Di Paolo, Michele De Tursi Tumor microenvironment and biomarkers P17 New immuno- histochemical markers for the differential diagnosis of atypical melanocytic lesions with uncertain malignant potential Laura Marra, Giosuè Scognamiglio, Monica Cantile, Margherita Cerrone, Fara De Murtas, Valeria Sorrentino, Anna Maria Anniciello, Gerardo Botti P18 Utility of simultaneous measurement of three serum tumor markers in melanoma patients Angela Sandru, Silviu Voinea, Eugenia Panaitescu, Madalina Bolovan, Adina Stanciu, Sabin Cinca P19 The significance of various cut-off levels of melanoma inhibitory activity in evaluation of cutaneous melanoma patients Angela Sandru, Silviu Voinea, Eugenia Panaitescu, Madalina Bolovan, Adina Stanciu, Sabin Cinca P20 The long noncoding RNA HOTAIR is associated to metastatic progression of melanoma and it can be identified in the blood of patients with advanced disease Chiara Botti, Giosuè Scognamiglio, Laura Marra, Gabriella Aquino, Rosaria Falcone, Annamaria Anniciello, Paolo Antonio Ascierto, Gerardo Botti, Monica Cantile Other P21 The effect of Sentinel Lymph Node Biopsy in melanoma mortality: timing of dissection Cristina Fortes, Simona Mastroeni, Alessio Caggiati, Francesca Passarelli, Alba Zappalà, Maria Capuano, Riccardo Bono, Maurizio Nudo, Claudia Marino, Paola Michelozzi P22 Epidemiological survey on related psychopathology in melanoma Valeria De Biasio, Vincenzo C. Battarra IMMUNOTHERAPY BRIDGE KEYNOTE SPEAKER PRESENTATIONS Immunotherapy beyond melanoma K19 Predictor of response to radiation and immunotherapy Silvia Formenti K20 Response and resistance to PD-1 pathway blockade: clues from the tumor microenvironment Maria Libera Ascierto, Tracee L. McMiller, Alan E. Berger, Ludmila Danilova, Robert A. Anders, George J. Netto, Haiying Xu, Theresa S. Pritchard, Jinshui Fan, Chris Cheadle, Leslie Cope, Charles G. Drake, Drew M. Pardoll, Janis M. Taube and Suzanne L. Topalian K21 Combination immunotherapy with autologous stem cell transplantation, protein immunization, and PBMC reinfusion in myeloma patients Sacha Gnjatic, Sarah Nataraj, Naoko Imai, Adeeb Rahman, Achim A. Jungbluth, Linda Pan, Ralph Venhaus, Andrew Park, Frédéric F. Lehmann, Nikoletta Lendvai, Adam D. Cohen, and Hearn J. Cho K22 Anti-cancer immunity despite T cell “exhaustion” Speiser Daniel Immunotherapy in oncology (I-O): data from clinical trial K23 The Checkpoint Inhibitors for the Treatment of Metastatic Non-small Cell Lung Cancer (NSCLC) Vera Hirsh
What problem does this paper attempt to address?
-
Future Perspectives in Melanoma Research
Paolo A. Ascierto,Sanjiv Agarwala,Gerardo Botti,Alessandra Cesano,Gennaro Ciliberto,Michael A. Davies,Sandra Demaria,Reinhard Dummer,Alexander M. Eggermont,Soldano Ferrone,Yang Xin Fu,Thomas F. Gajewski,Claus Garbe,Veronica Huber,Samir Khleif,Michael Krauthammer,Roger S. Lo,Giuseppe Masucci,Giuseppe Palmieri,Michael Postow,Igor Puzanov,Ann Silk,Stefani Spranger,David F. Stroncek,Ahmad Tarhini,Janis M. Taube,Alessandro Testori,Ena Wang,Jennifer A. Wargo,Cassian Yee,Hassane Zarour,Laurence Zitvogel,Bernard A. Fox,Nicola Mozzillo,Francesco M. Marincola,Magdalena Thurin
DOI: https://doi.org/10.1186/s12967-016-1070-y
IF: 8.44
2016-01-01
Journal of Translational Medicine
Abstract:The sixth “Melanoma Bridge Meeting” took place in Naples, Italy, December 1st–4th, 2015. The four sessions at this meeting were focused on: (1) molecular and immune advances; (2) combination therapies; (3) news in immunotherapy; and 4) tumor microenvironment and biomarkers. Recent advances in tumor biology and immunology has led to the development of new targeted and immunotherapeutic agents that prolong progression-free survival (PFS) and overall survival (OS) of cancer patients. Immunotherapies in particular have emerged as highly successful approaches to treat patients with cancer including melanoma, non-small cell lung cancer (NSCLC), renal cell carcinoma (RCC), bladder cancer, and Hodgkin’s disease. Specifically, many clinical successes have been using checkpoint receptor blockade, including T cell inhibitory receptors such as cytotoxic T-lymphocyte-associated antigen 4 (CTLA-4) and the programmed cell death-1 (PD-1) and its ligand PD-L1. Despite demonstrated successes, responses to immunotherapy interventions occur only in a minority of patients. Attempts are being made to improve responses to immunotherapy by developing biomarkers. Optimizing biomarkers for immunotherapy could help properly select patients for treatment and help to monitor response, progression and resistance that are critical challenges for the immuno-oncology (IO) field. Importantly, biomarkers could help to design rational combination therapies. In addition, biomarkers may help to define mechanism of action of different agents, dose selection and to sequence drug combinations. However, biomarkers and assays development to guide cancer immunotherapy is highly challenging for several reasons: (i) multiplicity of immunotherapy agents with different mechanisms of action including immunotherapies that target activating and inhibitory T cell receptors (e.g., CTLA-4, PD-1, etc.); adoptive T cell therapies that include tissue infiltrating lymphocytes (TILs), chimeric antigen receptors (CARs), and T cell receptor (TCR) modified T cells; (ii) tumor heterogeneity including changes in antigenic profiles over time and location in individual patient; and (iii) a variety of immune-suppressive mechanisms in the tumor microenvironment (TME) including T regulatory cells (Treg), myeloid derived suppressor cells (MDSC) and immunosuppressive cytokines. In addition, complex interaction of tumor-immune system further increases the level of difficulties in the process of biomarkers development and their validation for clinical use. Recent clinical trial results have highlighted the potential for combination therapies that include immunomodulating agents such as anti-PD-1 and anti-CTLA-4. Agents targeting other immune inhibitory (e.g., Tim-3) or immune stimulating (e.g., CD137) receptors on T cells and other approaches such as adoptive cell transfer are tested for clinical efficacy in melanoma as well. These agents are also being tested in combination with targeted therapies to improve upon shorter-term responses thus far seen with targeted therapy. Various locoregional interventions that demonstrate promising results in treatment of advanced melanoma are also integrated with immunotherapy agents and the 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 melanoma patients’ population. This meeting’s specific focus was on advances in immunotherapy and combination therapy for melanoma. The importance of understanding of melanoma genomic background for development of novel therapies and biomarkers for clinical application to predict the treatment response was an integral part of the meeting. The overall emphasis on biomarkers supports novel concepts toward integrating biomarkers into personalized-medicine approach for treatment 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. We also discussed the requirements for pre-analytical and analytical as well as clinical validation process as applied to biomarkers for cancer immunotherapy. The concept of the fit-for-purpose marker validation has been introduced to address the challenges and strategies for analytical and clinical validation design for specific assays.
-
Abstract 2486: Dissecting tumor-immune interaction in response and resistance to immune checkpoint blockade in metastatic melanoma
Giuseppe Tarantino,Yiwen He,Priyanka Solanky,Aaron Thorner,Tyler Aprati,Ryan Sullivan,Emily Robitschek,Timothy Blosser,Xiaowei Zhuang,Xiaowei Zhuang,Elliot Boblitt,Allison Frangieh,Marta Holovatska,Aleigha Lawless,Michael Manos,Kathleen Pfaff,Karla Helvie,Tatyana Sharova,Dennie Frederick,James Liam Fahey,Diego Villamarin,Sami Farhi,Scott Rodig,Bruce Johnson,Alex K. Shalek,Eliezer Van Allen,Stephen Hodi,Genevieve M. Boland,David Liu
DOI: https://doi.org/10.1158/1538-7445.am2024-2486
IF: 11.2
2024-03-22
Cancer Research
Abstract:Abstract Background: Immune checkpoint blockade (ICB) has revolutionized the treatment and prognosis of various cancers, including melanoma. Nevertheless, intrinsic or acquired resistance is common, and the interactions between tumor-intrinsic features and the microenvironment underlying these resistances remain unclear for most patients. Methods: In this study, we developed a framework to analyze response and resistance, both intrinsic and acquired, via tumor-intrinsic programs, immune features, and tumor-stromal-immune interactions in a standardized, uniformly processed, and deeply clinically annotated cohort of metastatic melanoma patients (n=61) treated with ICB as part of the NCI funded human tumor atlas network (HTAN) initiative. From the same tumor samples (pre-treatment (n=33) and post-progression (n=28)), we conducted bulk whole-exome sequencing (WES), single-nucleus RNA sequencing, and for a subset of the samples high-resolution spatial imaging (including protein mIHC, CODEX, and transcriptomics MERFISH). Standardized processing and data pipelines allowed for integration of genomic, transcriptomic, and spatial features to elucidate shared tumor and microenvironmental states and their relationships with resistance. Results: Our preliminary analysis on a subset of 28 samples identified an enrichment of B cells, plasma cells, and T follicular helper-like cells in the tumor microenvironment (TME) of patients sensitive to treatment. The TME of non-responders was predominantly characterized by macrophage/monocyte cell populations. Post-treatment responder samples showed an enrichment of CD4+ T cells. Genomic heterogeneity, assessed through WES, was confirmed as a feature of intrinsic resistance across different ICB treatment settings, while ploidy was associated with treatment response in pre-treatment, ipilimumab-naïve patients treated with PD-1 inhibitors. Comparing the immune compartments of samples with high and low heterogeneity revealed NK cell enrichment in samples with a high proportion of subclonal mutations. Conclusions and Future Directions: Overall, our work provides a high-resolution understanding of the tumor-stromal-immune interaction in metastatic melanoma, shedding light on the factors contributing to therapy response and resistance, and contributing a repository of data from deeply clinically annotated, multimodally characterized and uniformly processed patient samples. Ongoing spatial analysis will further enhance our understanding of these compartments at a spatial level. The results of our study have the potential to guide more personalized and effective treatment strategies for metastatic melanoma in the future. Citation Format: Giuseppe Tarantino, Yiwen He, Priyanka Solanky, Aaron Thorner, Tyler Aprati, Ryan Sullivan, Emily Robitschek, Timothy Blosser, Xiaowei Zhuang, Xiaowei Zhuang, Elliot Boblitt, Allison Frangieh, Marta Holovatska, Aleigha Lawless, Michael Manos, Kathleen Pfaff, Karla Helvie, Tatyana Sharova, Dennie Frederick, James Liam Fahey, Diego Villamarin, Sami Farhi, Scott Rodig, Bruce Johnson, Alex K. Shalek, Eliezer Van Allen, Stephen Hodi, Genevieve M. Boland, David Liu. Dissecting tumor-immune interaction in response and resistance to immune checkpoint blockade in metastatic melanoma [abstract]. In: Proceedings of the American Association for Cancer Research Annual Meeting 2024; Part 1 (Regular Abstracts); 2024 Apr 5-10; San Diego, CA. Philadelphia (PA): AACR; Cancer Res 2024;84(6_Suppl):Abstract nr 2486.
oncology
-
Abstract B141: Combining the telomerase peptide cancer vaccine UV1 with CTLA-4 blockade in patients with metastatic malignant melanoma: Proof of principle and early clinical reports from a phase I/IIa trial
G. Kvalheim,T. Guren,E. Aamdal,Øyvind Arnesen,S. Aamdal,G. Gaudernack,J. Kyte,E. Suso
DOI: https://doi.org/10.1158/2326-6074.CRICIMTEATIAACR15-B141
Abstract:The purpose of this clinical phase I/IIa, open label, single arm, interventional study (EudraCT No 2013-005582-39) is to investigate the safety, clinical and immunological responses of a therapeutic telomerase peptide vaccine, UV1, combined with ipilimumab in patients with metastatic melanoma (MM). The CTLA-4 antibody ipilimumab produces long term survival benefits in ca.20% of patients with MM. The mechanism of action of ipilimumab suggests that combinations with therapies inducing tumor-specific immune responses, such as vaccines, may lead to additive and even synergistic anti-tumor activity. We hypothesized that a telomerase peptide-based vaccine in combination with ipilimumab would improve therapeutic efficacy in patients with MM. Telomerase promotes elongation of telomeres after each cell division, representing the key enzymatic process in preventing telomere shortening. It is responsible for human cell immortalization and cancer pathogenesis, and is present in 85-90% of cancer tissues. Thus, telomerase-based immunotherapy has been investigated in several tumor types. Available blood samples from long term cancer survivors previously treated with different types of human telomerase reverse transcriptase subunit (hTERT) vaccines identified a new set of peptide epitopes. T cell responses to these peptides form spontaneously after vaccination by epitope spreading and are found only in patients with remarkable clinical courses, suggesting a role in tumor eradication. This led to the development of UV1, a therapeutic cancer vaccine consisting of a mixture of 3 synthetic peptides representing naturally occurring fragments of human hTERT. Ongoing phase I/IIa trials with UV1 in non-small-cell lung cancer and prostate cancer (EudraCT No 2012-001852-20 and 2012-002411-26, respectively) have shown potent, durable T cell responses against UV1 peptides and low toxicity (unpublished data). In the present protocol, patients with a histologically or cytologically confirmed diagnosis of unresectable AJCC stage III/IV MM, ECOG 0-1 and an adequate renal, hepatic and hematological function are eligible. Any previous treatment is allowed. Patients with active brain metastases, a history of autoimmune disease, splenic surgery or irradiation, allogenic stem cell transplantation, known hypersensitivity to the investigational products, uncontrolled infectious disease, pregnant or breastfeeding, will be excluded. Intradermal UV1 vaccines of 300 mcg and 75 mcg GM-CSF are administered 7, 5, and 3 days before and 11 days after the first dose of ipilimumab, then 3 days before every dose of ipilimumab, and subsequently every 4th week until week 48 or treatment cessation. Ipilimumab 3 mg/kg is given IV every 3rd week for a total of 4 doses. Each patient will be followed up 5 weeks after the completion of the last study treatment. Adverse events are recorded in coherence with CTCAE vs. 4.0. Tumor response is evaluated according to RECIST vs.1.1. Immune responses against UV1 peptides are monitored by standard immunoassays. Potential predictive biomarkers will be explored in an extensive program involving sequential tumor biopsies, DNA sequencing and mRNA expression. The first patient was enrolled 16 January 2015, estimated completed recruitment by one year. 3 out of 20 patients have been recruited so far. Data from the first 3 patients until week 16 will be reported. Citation Format: Elin Aamdal, Tormod Kyrre Guren, Else Marit Inderberg Suso, Gunnar Kvalheim, Jon Amund Kyte, Oyvind Kongstun Arnesen, Steinar Aamdal, Gustav Gaudernack. Combining the telomerase peptide cancer vaccine UV1 with CTLA-4 blockade in patients with metastatic malignant melanoma: Proof of principle and early clinical reports from a phase I/IIa trial. [abstract]. In: Proceedings of the CRI-CIMT-EATI-AACR Inaugural International Cancer Immunotherapy Conference: Translating Science into Survival; September 16-19, 2015; New York, NY. Philadelphia (PA): AACR; Cancer Immunol Res 2016;4(1 Suppl):Abstract nr B141.
Medicine
-
Interaction of Molecular Alterations with Immune Response in Melanoma
Robert A. Szczepaniak Sloane,Vancheswaran Gopalakrishnan,Sangeetha M. Reddy,Xue Zhang,Alexandre Reuben,Jennifer A. Wargo
DOI: https://doi.org/10.1002/cncr.30681
IF: 6.9209
2017-01-01
Cancer
Abstract:Major advances have been made in melanoma treatment with the use of molecularly targeted therapies and immunotherapies, and numerous regimens are now approved by the US Food and Drug Administration for patients with stage IV disease. However, therapeutic resistance remains an issue to both classes of agents, and reliable biomarkers of therapeutic response and resistance are lacking. Mechanistic insights are being gained through preclinical studies and translational research, offering potential strategies to enhance responses and survival in treated patients. A comprehensive understanding of the immune effects of common mutations at play in melanoma is critical, as is an appreciation of the molecular mechanisms contributing to therapeutic resistance to immunotherapy. These mechanisms and the interplay between them are discussed herein. (c) 2017 American Cancer Society.
-
Abstract 6397: Immune profiling for improving responsiveness to ipilimumab plus nivolumab checkpoint blockade treatment in patients with metastatic melanoma
Joanna Baginska,Jiajia Chen,Giuseppe Tarantino,Anita Giobbie-Hurder,Jason L. Weirather,Mariano Severgnini,Michael Manos,Kelly Burke,Janice D. Russell,Martha Brainard,Elizabeth Gabriel,Ryan Brennick,Matthew Nazzaro,Emma Hathaway,Marta Holovatska,Claire Manuszak,Srinika Ranashinge,David Liu,F. Stephen Hodi
DOI: https://doi.org/10.1158/1538-7445.am2024-6397
IF: 11.2
2024-03-28
Cancer Research
Abstract:Background:aCTLA-4 and aPD-1 combination therapy has significantly improved clinical outcomes in patients with metastatic melanoma, with 50%-60% of patients responding to treatment, but predictors of response and treatment induced immunological effects are poorly characterized. Identifying circulating immune populations as biomarkers could reveal crucial insights into the mechanisms driving treatment success. Methods:Using unbiased single-cell profiling (with cytometry by Time-Of-Flight, CyTOF), we characterized the systemic immune compartments of over 250 peripheral blood mononuclear samples from 89 patients with advanced melanoma before and during combination therapy. We evaluated the association between circulating immune populations and clinical outcome using nonparametric Mann-Whitney tests, and investigated the prognostic values of these features using survival analysis. Additionally, we assessed treatment-induced immunological effects, providing insights into the dynamic changes within the immune system during therapy. Results:We characterized circulating T cells, NK cells, B cells, and monocytes from peripheral blood. Following combination therapy, there was a shift from naïve and central memory T cells to effector populations in both CD4+ and CD8+ T cells compartments. There was also a broad activation observed in both adaptive and innate populations, characterized by an elevation of multiple co-stimulatory and co-inhibitory molecules. Lower CD8+ NK cell populations prior to combination therapy were predictive of durable clinical benefit (DCB). Pre-treatment measurements of Tregs were not predictive of outcome, but higher Tregs one month after starting therapy, as well as increases of Tregs from pre to post treatment, predicted better progression free survival (PFS) and overall survival (OS). Starting from early on treatment, higher "exhausted-like" CD8+ T cells, characterized by positive expression of at least two co-inhibitory markers, is associated with resistance. Conclusions:aCTLA-4 and aPD-1 combination therapy led to a longitudinal shift from naïve and central memory to effector phenotypes in T cells, along with a broad activation in both adaptive and innate populations. Within innate immune compartment, favorable clinical outcome was associated with lower CD8+ NK cells, capable of regulating proliferation and activation of CD4+ T cells. Interestingly, one month after the start of the therapy, the increase in Tregs and a decrease in 'exhausted-like' CD8+ T populations are associated with a better response. Collectively, these analyses suggest potential treatment-specific correlates of efficacy and may enable biomarker-selected patient populations in future. Citation Format: Joanna Baginska, Jiajia Chen, Giuseppe Tarantino, Anita Giobbie-Hurder, Jason L. Weirather, Mariano Severgnini, Michael Manos, Kelly Burke, Janice D. Russell, Martha Brainard, Elizabeth Gabriel, Ryan Brennick, Matthew Nazzaro, Emma Hathaway, Marta Holovatska, Claire Manuszak, Srinika Ranashinge, David Liu, F. Stephen Hodi. Immune profiling for improving responsiveness to ipilimumab plus nivolumab checkpoint blockade treatment in patients with metastatic melanoma [abstract]. In: Proceedings of the American Association for Cancer Research Annual Meeting 2024; Part 1 (Regular s); 2024 Apr 5-10; San Diego, CA. Philadelphia (PA): AACR; Cancer Res 2024;84(6_Suppl) nr 6397.
oncology
-
Therapeutic Advancements Across Clinical Stages in Melanoma, With a Focus on Targeted Immunotherapy
Claudia Trojaniello,Jason J Luke,Paolo A Ascierto
DOI: https://doi.org/10.3389/fonc.2021.670726
2021-06-10
Abstract:Melanoma is the most fatal skin cancer. In the early stages, it can be safely treated with surgery alone. However, since 2011, there has been an important revolution in the treatment of melanoma with new effective treatments. Targeted therapy and immunotherapy with checkpoint inhibitors have changed the history of this disease. To date, more than half of advanced melanoma patients are alive at 5 years; despite this breakthrough, approximately half of the patients still do not respond to treatment. For these reasons, new therapeutic strategies are required to expand the number of patients who can benefit from immunotherapy or combination with targeted therapy. Current research aims at preventing primary and acquired resistance, which are both responsible for treatment failure in about 50% of patients. This could increase the effectiveness of available drugs and allow for the evaluation of new combinations and new targets. The main pathways and molecules under study are the IDO inhibitor, TLR9 agonist, STING, LAG-3, TIM-3, HDAC inhibitors, pegylated IL-2 (NKTR-214), GITR, and adenosine pathway inhibitors, among others (there are currently about 3000 trials that are evaluating immunotherapeutic combinations in different tumors). Other promising strategies are cancer vaccines and oncolytic viruses. Another approach is to isolate and remove immune cells (DCs, T cells, and NK cells) from the patient's blood or tumor infiltrates, add specific gene fragments, expand them in culture with growth factors, and re-inoculate into the same patient. TILs, TCR gene transfer, and CAR-T therapy follow this approach. In this article, we give an overview over the current status of melanoma therapies, the clinical rationale for choosing treatments, and the new immunotherapy approaches.
-
Clinical Models to Define Response and Survival With Anti–PD-1 Antibodies Alone or Combined With Ipilimumab in Metastatic Melanoma
Inês Pires da Silva,Tasnia Ahmed,Jennifer L. McQuade,Caroline A. Nebhan,John J. Park,Judith M. Versluis,Patricio Serra-Bellver,Yasir Khan,Tim Slattery,Honey K. Oberoi,Selma Ugurel,Lauren E. Haydu,Rudolf Herbst,Jochen Utikal,Claudia Pföhler,Patrick Terheyden,Michael Weichenthal,Ralf Gutzmer,Peter Mohr,Rajat Rai,Jessica L. Smith,Richard A. Scolyer,Ana M. Arance,Lisa Pickering,James Larkin,Paul Lorigan,Christian U. Blank,Dirk Schadendorf,Michael A. Davies,Matteo S. Carlino,Douglas B. Johnson,Georgina V. Long,Serigne N. Lo,Alexander M. Menzies
DOI: https://doi.org/10.1200/jco.21.01701
IF: 45.3
2022-04-01
Journal of Clinical Oncology
Abstract:PURPOSE Currently, there are no robust biomarkers that predict immunotherapy outcomes in metastatic melanoma. We sought to build multivariable predictive models for response and survival to anti-programmed cell death protein 1 (anti–PD-1) monotherapy or in combination with anticytotoxic T-cell lymphocyte-4 (ipilimumab [IPI]; anti–PD-1 ± IPI) by including routine clinical data available at the point of treatment initiation. METHODS One thousand six hundred forty-four patients with metastatic melanoma treated with anti–PD-1 ± IPI at 16 centers from Australia, the United States, and Europe were included. Demographics, disease characteristics, and baseline blood parameters were analyzed. The end points of this study were objective response rate (ORR), progression-free survival (PFS), and overall survival (OS). The final predictive models for ORR, PFS, and OS were determined through penalized regression methodology (least absolute shrinkage and selection operator method) to select the most significant predictors for all three outcomes (discovery cohort, N = 633). Each model was validated internally and externally in two independent cohorts (validation-1 [N = 419] and validation-2 [N = 592]) and nomograms were created. RESULTS The final model for predicting ORR (area under the curve [AUC] = 0.71) in immunotherapy-treated patients included the following clinical parameters: Eastern Cooperative Oncology Group Performance Status, presence/absence of liver and lung metastases, serum lactate dehydrogenase, blood neutrophil-lymphocyte ratio, therapy (monotherapy/combination), and line of treatment. The final predictive models for PFS (AUC = 0.68) and OS (AUC = 0.77) included the same variables as those in the ORR model (except for presence/absence of lung metastases), and included presence/absence of brain metastases and blood hemoglobin. Nomogram calculators were developed from the clinical models to predict outcomes for patients with metastatic melanoma treated with anti–PD-1 ± IPI. CONCLUSION Newly developed combinations of routinely collected baseline clinical factors predict the response and survival outcomes of patients with metastatic melanoma treated with immunotherapy and may serve as valuable tools for clinical decision making.
oncology
-
Biomarkers Predictive of Survival and Response to Immune Checkpoint Inhibitors in Melanoma
Emanuelle M. Rizk,Angelina M. Seffens,Megan H. Trager,Michael R. Moore,Larisa J. Geskin,Robyn D. Gartrell-Corrado,Winston Wong,Yvonne M. Saenger
DOI: https://doi.org/10.1007/s40257-019-00475-1
2019-10-10
American Journal of Clinical Dermatology
Abstract:Immunotherapy has revolutionized the treatment of melanoma. Targeting of the immune checkpoints cytotoxic T-lymphocyte-associated protein 4 and programmed cell death protein 1 has led to improved survival in a subset of patients. Unfortunately, the use of immune checkpoint inhibitors is associated with significant side effects and many patients do not respond to treatment. Thus, there is an urgent need both for prognostic biomarkers to estimate risk and for predictive biomarkers to determine which patients are likely to respond to therapy. In this review, prognostic and predictive biomarkers that are an active area of research are outlined. Of note, certain transcriptomic signatures are already used in the clinic, albeit not routinely, to prognosticate patients. In the predictive setting, programmed cell death protein ligand 1 expression has been shown to correlate with benefit but is not precise enough to be used as an exclusionary biomarker. Future investigation will need to focus on biomarkers that are easily reproducible, cost effective, and accurate. The use of readily available clinical material, such as serum or hematoxylin and eosin-stained images, may offer one such path forward.
dermatology
-
Abstract 6550: Biopsy analysis of trial S1616: Ipilimumab plus nivolumab versus ipilimumab alone in patients with anti-PD-1 refractory melanoma
Katie M. Campbell,Zaid Bustami,Daniel G. Chen,Egmidio Medina,Cynthia R. Gonzalez,Nataly Naser Aldeen,Ignacio Baselga-Carretero,Agustin Vega-Crespo,Jessica Maxey,Jia M. Chen,Lawrence F. Kuklinski,Kari L. Kendra,Bartosz Chmielowski,Thach-Giao Truong,Nikhil I. Khushalani,Frances Collichio,Alexandra Ikeguchi,Adrienne I. Victor,Kim Margolin,Jeffrey A. Sosman,Sapna P. Patel,Siwen Hu-Lieskovan,James Moon,Shay Bellasea,Daniel K. Wells,Christine N. Spencer,Marshall A. Thompson,Michael Wu,Philip O. Scumpia,Ari VanderWalde,Antoni Ribas
DOI: https://doi.org/10.1158/1538-7445.am2024-6550
IF: 11.2
2024-03-28
Cancer Research
Abstract:Background: The phase II randomized trial S1616 (NCT03033576) showed that patients with melanoma refractory to anti-PD-1-based therapy had improved progression free survival (HR=0.63, p=0.037) and objective response (28% vs 9%) to the combination of ipilimumab with nivolumab compared to ipilimumab. Here, we report molecular and spatial proteomic features of biopsies collected from patients on S1616, both prior to and during therapy. Methods: Biopsies collected from patients from both arms at baseline (N=68 patients total) and early on-therapy (N=51; 43 with paired timepoints) were analyzed by whole exome sequencing (n=185 samples), RNA sequencing (n=105), histopathologic staining (n=149), and multiplexed ion beam imaging (n=45). Multiple biopsies were available for some patients. Mutations, gene expression, and tumor microenvironment were compared across timepoints and response to combination (N=18 responders [CR/PR], N=44 nonresponders [SD/PD]). Results: Baseline biopsies from patients responsive to combination had increased expression of genes (n=482, FDR<0.05) associated with coagulation and complement, fatty acid metabolism, oxidative phosphorylation, hypoxia, and interferon gamma response gene sets (FDR<0.05), compared to nonresponsive biopsies. Baseline biopsies from responders also had low levels of effector CD8 T cells (PD1+, TIM-3+, GZMB+, Ki67+) colocalized with tumor cells and myeloid populations expressing higher levels of MHC Class II. On-therapy biopsies from responders showed decreased detection of driver mutations by genomics, reduced gene expression of pathways enriched at baseline (oxidative phosphorylation, complement) by transcriptomics, and increased CD8 T cell to tumor cell ratios by histopathology, supporting observations of tumor regression. On-therapy biopsies from responders also had increased gene expression of genes related to inflammatory cytokine signaling. This correlated with increased proportions of effector CD8 T cells, compared to paired baseline or nonresponding biopsies, and increased organization of nonactivated CD8 T cells near mature endothelial structures (SMA+, CD31+ regions) adjacent to tumor. Biopsies from nonresponders did not demonstrate these dynamics and instead contained exhausted CD8 T cells (PD1+, TIM-3+, granzyme B-, Ki67-) colocalized with FOXP3+, CD4+ Tregs and CD163+, PD-L1+ M2 macrophages, both at baseline and on-therapy. Conclusion: In patients with melanoma refractory to anti-PD-1, addition of anti-CTLA-4 facilitates tumor-reactive CD8 T-cell infiltration and decreased suppressor cell dynamics, resulting in regression of some tumors with distinct transcriptome features. Conversely, biopsies from patients whose tumors progress on combination therapy lack expression of metabolic pathways and show CD8 T-cells restricted in proximity to M2 macrophages and Tregs. Citation Format: Katie M. Campbell, Zaid Bustami, Daniel G. Chen, Egmidio Medina, Cynthia R. Gonzalez, Nataly Naser Aldeen, Ignacio Baselga-Carretero, Agustin Vega-Crespo, Jessica Maxey, Jia M. Chen, Lawrence F. Kuklinski, Kari L. Kendra, Bartosz Chmielowski, Thach-Giao Truong, Nikhil I. Khushalani, Frances Collichio, Alexandra Ikeguchi, Adrienne I. Victor, Kim Margolin, Jeffrey A. Sosman, Sapna P. Patel, Siwen Hu-Lieskovan, James Moon, Shay Bellasea, Daniel K. Wells, Christine N. Spencer, Marshall A. Thompson, Michael Wu, Philip O. Scumpia, Ari VanderWalde, Antoni Ribas. Biopsy analysis of trial S1616: Ipilimumab plus nivolumab versus ipilimumab alone in patients with anti-PD-1 refractory melanoma [abstract]. In: Proceedings of the American Association for Cancer Research Annual Meeting 2024; Part 1 (Regular s); 2024 Apr 5-10; San Diego, CA. Philadelphia (PA): AACR; Cancer Res 2024;84(6_Suppl) nr 6550.
oncology
-
Predicting response to checkpoint inhibitors in melanoma beyond PD-L1 and mutational burden
Carl Morrison,Sarabjot Pabla,Jeffrey M Conroy,Mary K Nesline,Sean T Glenn,Devin Dressman,Antonios Papanicolau-Sengos,Blake Burgher,Jonathan Andreas,Vincent Giamo,Moachun Qin,Yirong Wang,Felicia L Lenzo,Angela Omilian,Wiam Bshara,Matthew Zibelman,Pooja Ghatalia,Konstantin Dragnev,Keisuke Shirai,Katherine G Madden,Laura J Tafe,Neel Shah,Deepa Kasuganti,Luis de la Cruz-Merino,Isabel Araujo,Yvonne Saenger,Margaret Bogardus,Miguel Villalona-Calero,Zuanel Diaz,Roger Day,Marcia Eisenberg,Steven M Anderson,Igor Puzanov,Lorenzo Galluzzi,Mark Gardner,Marc S Ernstoff
DOI: https://doi.org/10.1186/s40425-018-0344-8
2018-05-09
Abstract:Background: Immune checkpoint inhibitors (ICIs) have changed the clinical management of melanoma. However, not all patients respond, and current biomarkers including PD-L1 and mutational burden show incomplete predictive performance. The clinical validity and utility of complex biomarkers have not been studied in melanoma. Methods: Cutaneous metastatic melanoma patients at eight institutions were evaluated for PD-L1 expression, CD8+ T-cell infiltration pattern, mutational burden, and 394 immune transcript expression. PD-L1 IHC and mutational burden were assessed for association with overall survival (OS) in 94 patients treated prior to ICI approval by the FDA (historical-controls), and in 137 patients treated with ICIs. Unsupervised analysis revealed distinct immune-clusters with separate response rates. This comprehensive immune profiling data were then integrated to generate a continuous Response Score (RS) based upon response criteria (RECIST v.1.1). RS was developed using a single institution training cohort (n = 48) and subsequently tested in a separate eight institution validation cohort (n = 29) to mimic a real-world clinical scenario. Results: PD-L1 positivity ≥1% correlated with response and OS in ICI-treated patients, but demonstrated limited predictive performance. High mutational burden was associated with response in ICI-treated patients, but not with OS. Comprehensive immune profiling using RS demonstrated higher sensitivity (72.2%) compared to PD-L1 IHC (34.25%) and tumor mutational burden (32.5%), but with similar specificity. Conclusions: In this study, the response score derived from comprehensive immune profiling in a limited melanoma cohort showed improved predictive performance as compared to PD-L1 IHC and tumor mutational burden.
-
Immunotherapy in the Treatment of Metastatic Melanoma: Current Knowledge and Future Directions
Massimo Ralli,Andrea Botticelli,Irene Claudia Visconti,Diletta Angeletti,Marco Fiore,Paolo Marchetti,Alessandro Lambiase,Marco de Vincentiis,Antonio Greco
DOI: https://doi.org/10.1155/2020/9235638
IF: 4.4929
2020-06-28
Journal of Immunology Research
Abstract:Melanoma is one of the most immunologic malignancies based on its higher prevalence in immune-compromised patients, the evidence of brisk lymphocytic infiltrates in both primary tumors and metastases, the documented recognition of melanoma antigens by tumor-infiltrating T lymphocytes and, most important, evidence that melanoma responds to immunotherapy. The use of immunotherapy in the treatment of metastatic melanoma is a relatively late discovery for this malignancy. Recent studies have shown a significantly higher success rate with combination of immunotherapy and chemotherapy, radiotherapy, or targeted molecular therapy. Immunotherapy is associated to a panel of dysimmune toxicities called immune-related adverse events that can affect one or more organs and may limit its use. Future directions in the treatment of metastatic melanoma include immunotherapy with anti-PD1 antibodies or targeted therapy with BRAF and MEK inhibitors.
immunology
-
Abstract NG04: Immune evasion mechanisms in melanoma resistant to targeted- and/or immunotherapy
Willy Hugo,Lu Sun,Marco Piva,Shirley Lomeli,Chunying Song,Antoni Ribas,Roger S. Lo
DOI: https://doi.org/10.1158/1538-7445.am2018-ng04
2018-07-01
Immunology
Abstract:Abstract Despite recent advances in mutation- targeted and immune therapies for melanoma, prolonged patient survival is still hampered by therapeutic resistance. For MAPKi, acquired resistance is common. For example, a majority of V600E/K mutant BRAF metastatic melanomas treated with MAPK inhibitor (MAPKi) will acquire resistance within 1-2 years of the treatment initiation, despite BRAF inhibitors (BRAFi) and their combination with MEK inhibitors (BRAFi+MEKi) being remarkably active initially. Our work and others' have shown that immune modulation by MAPKi may contribute to the in vivo anti-tumor activities of this oncogene-targeted approach. Importantly, my own recent study has shown that around half of all acquired or late MAPKi resistant melanoma display profound CD8 T-cell depletion and exhaustion, which were tightly associated with reduced antigen-presentation. Immunotherapies using antibodies against the T cell checkpoints, CTLA-4 and PD-1, have been shown to enhance the ability of endogenous T cells to destroy malignant cells in a variety of cancers, including melanoma. When successful, these therapies can result durable tumor control. However, the majority of patients do not respond to such therapies. Multiple studies including ours have delineated multiple factors limiting the efficacy of single agent anti-CTLA-4 or anti-PD-1 treatments, including the lack of tumor infiltration by T cells, insensitivity to IFN-gamma signaling and the activation of immune-suppressive processes represented by Innate anti-PD-1 RESistance (IPRES) signatures. Functionally, IPRES signatures include the downstream genes of potent immune-suppressive pathways and wound-healing processes linked to suppression and exclusion of T cells (e.g., angiogenesis, extracellular matrix reorganization, mesenchymal/invasive transition). Unsupervised gene signature enrichment clustering revealed four subclasses with combinations of high/low enrichment scores of interferon (IFN)/T-cell inflammation and of IPRES related signatures in both our dataset and the TCGA melanoma. The cellular processes represented by the four subsets significantly resemble those induced during wound healing where: 1) the IFN-high stage matches the early, acute inflammation stage and 2) the IPRES-high stage matches the stage of neovascularization, extracellular matrix remodeling and suppression of inflammation. Importantly, the patients from different subclasses have statistically different overall survival where the IFN-high/IPRES-low group showed the best survival and IFN-low/IPRES-low group was the worst. In murine mutant BRAF melanoma YUMM1.7, regressing/responding tumors on the 6th day of BRAFi treatment showed IFN-high/IPRES-low pattern with a significant infiltration of T cells and other immune cell populations into the tumor. The residual tumors on the 15th day showed IFN-high/IPRES-high pattern and showed further immune infiltration. By the time of acquired resistance to BRAFi, however, all tumors were in the IFN-low/ IPRES-high stage and there was no or minimal immune infiltration in the tumor. We observed that MAPKi-induced an IFN-high/IPRES-high state in the residual melanoma tumors from patients undergoing MAPKi treatment. Given the immune suppressive nature of IPRES, we hypothesized that its activation may cause the observed intratumoral immune (T-cell, in particular) dysfunction and depletion. Indeed, combination of MAPKi with immune checkpoint blockade (ICB) therapy targeting the PD-1 only works when the ICB is administered before the IFN-high/IPRES-high stage. Also intriguing is the finding that IPRES was induced in vitro along with a tumor immune phenotype switch MAPKi resistant melanoma cell lines, suggesting, a tumor cell-intrinsic process. We found that IPRES signatures include potent immune-suppressive pathways like the VEGF and TGFB pathways. TGFB signaling is known to regulate T-cell inflammation and promote T-cell tolerance by downregulating TCR signaling. TGFB ligands (TGFB1/2/3 and ACTIVIN-A/B) are normally secreted by immune cells including T-cells, M2-macrophages and endothelial cells during the inflammation resolution phase of wound healing. In our data and the TCGA melanoma, we observed higher mRNA and protein expression of TGFB ligands in the IPRES-high melanoma subgroups and in tumors treated with MAPKi. We postulate that high levels of TGFB ligands, from tumor and stromal sources, could counteract the T-cell anti-tumor immunity. Of interest, a study has shown some efficacy of targeting the TGFB pathway by fresolimumab (a human monoclonal antibody against TGFB1-3) in melanoma (Clinical Trial ID: NCT0035646017). Another study currently combines nivolumab (anti-PD-1) with galunisertib (a TGFBR type I/II small molecule inhibitor) in multiple solid tumors (NCT02423343). In relation to the VEGF pathway, we observed an anti-correlation between the mRNA levels of VEGFA and CD8A in both our MAPKi and the TCGA melanoma cohorts. We also observed that VEGFA mRNA and its protein level were expressed higher in the non-IFN-/T-cell inflamed subclasses. Several publications had shown that tumors that were non-responsive to anti-PD-1 have higher VEGFA levels associated with either PTEN loss or beta-catenin activation. Tumor-secreted VEGFA was reported to inhibit T-cell diapedesis from the vasculature, and thus infiltration into the tumor bed. Our observations are thus in line with such a role of VEGFA in blocking T-cell homing into the tumor. Consistently, a recent report showed that anti-VEGFA antibody pre-treatment followed by co-treatment with aPD-L1 improved treatment responses among renal cell carcinoma (RCC) patients. The authors reported increased tumor-specific T-cell trafficking into the tumor with the dual treatment. An earlier study combining anti-CTLA-4 with anti-VEGFA also showed similar result. Thus, anti-VEGFA combined with anti-PD-1/L1 may have a role in addressing non-T-cell inflamed melanoma with innate/acquired aPD-1 resistance or acquired MAPKi-resistance. Thus, given the clinical availability of both TGFB and VEGFA antagonists, we are testing the efficacy of combinatorial MAPKi and anti-TGFB/VEGFA to improve T-cell mediated, antitumor immunity through suppression of IPRES. Finally, our recent study showed that MAPKi-treated, regressing melanoma already evolve epigenetic alterations, temporally far ahead of any clinical evidence of disease progression. However, it was not clear if the observed transition into an IPRES-high, immune-suppressive state in these melanoma has any epigenetic basis. In parallel studies in cell lines, we characterized two groups of melanomas derived from continuous treatment with BRAFi or BRAFi+MEKi: MAPK-redundant (Rr) vs. MAPK-reactivated (Ra). The Ra lines could be resensitized to additional MAPK inhibitors while the Rr lines could not (due to MAPK-independent growth and surival). Intriguingly, only the Rr-lines showed enrichments of the IPRES signatures. Importantly, we noted that chromatin enhancer regions (marked by significant H3K27ac peaks) of the Rr lines were correlated with more invasive melanomas. The same invasive melanoma enhancer profile can also be observed in the patient-derived MAPKi-on-treatment tumors. By analyzing the actual mRNA expression changes of the genes proximal to the invasive melanoma specific H3K27ac peaks, we found that the upregulated genes were enriched with IPRES-related processes. This implies that the tumor cell-intrinsic up-expression of IPRES-related genes is regulated at the epigenetic level. In summary, we anticipate that our study will lead to novel insights into the determinants and/or bottlenecks of effective and durable anti-tumor responses by tumor-specific T-cells in order to pave the way to a new combination therapies in human melanoma patients. Citation Format: Willy Hugo, Lu Sun, Marco Piva, Shirley Lomeli, Chunying Song, Antoni Ribas, Roger S. Lo. Immune evasion mechanisms in melanoma resistant to targeted- and/or immunotherapy [abstract]. In: Proceedings of the American Association for Cancer Research Annual Meeting 2018; 2018 Apr 14-18; Chicago, IL. Philadelphia (PA): AACR; Cancer Res 2018;78(13 Suppl):Abstract nr NG04.
immunology
-
Immunomodulating antibodies in the treatment of metastatic melanoma: the experience with anti-CTLA-4, anti-CD137, and anti-PD1
Ester Simeone,Paolo A Ascierto
DOI: https://doi.org/10.3109/1547691X.2012.678021
Abstract:Clinical activity of anti-CTLA-4 (cytotoxic T-lymphocyte antigen-4) monoclonal antibodies (mAb) has changed the approaches for the treatment of cancer in terms of patterns of response, duration of response, and adverse event profiles. In fact, antibodies that block the interaction of CTLA-4 with its ligands B7.1 and B7.2 can enhance immune responses, including anti-tumor immunity. Two recent studies using ipilimumab (an anti-CTLA-4 mAb) demonstrated improvements in overall survival in the treatment of advanced melanoma. These studies utilized two different schedules of treatment in different patient categories (first and second line of treatment). However, the results were quite similar despite the different dosage used and the combination with dacarbazine in the first line treatment. Ongoing clinical studies will establish the efficacy of ipilimumab as monotherapy or in combination with other drugs for the treatment of metastatic melanoma and a variety of other cancers. Other antibodies, such as CD137 agonists and PD-1 antagonists, are currently in various stages of pre-clinical and clinical development. Agonist antibodies directed against CD137 (4-1BB) on the surface of antigen-primed T-lymphocytes increase tumor immunity that is curative against some transplantable murine tumors. Programmed death-1 (PD1) is a surface molecule delivering inhibitory signals important to maintain T-cell functional silence against their cognate antigens. Interference with PD1 or its ligand PD-L1 (B7-H1) increases anti-tumor immunity. As a result, human mAbs anti-PD1 and anti-PD-L1 are under clinical development. This paper reviews recent studies in the treatment of advanced melanoma with these types of monoclonal antibodies. Ipilimumab can be considered a cornerstone of a new era in melanoma treatment. However, the aim is to optimize the therapy with anti-CTLA-4 antibodies to define the best schedule for next combination regimens (other immunomodulatory antibodies, BRAF/MEK inhibitors, vaccines, etc.) that represent the natural evolution of future melanoma therapy.
-
Antibodies as biomarker candidates for response and survival to checkpoint inhibitors in melanoma patients
Mirjam Fässler,Stefan Diem,Joanna Mangana,Omar Hasan Ali,Fiamma Berner,David Bomze,Sandra Ring,Rebekka Niederer,Cristina del Carmen Gil Cruz,Christian Ivan Pérez Shibayama,Michal Krolik,Marco Siano,Markus Joerger,Mike Recher,Lorenz Risch,Sabine Güsewell,Martin Risch,Daniel E. Speiser,Burkhard Ludewig,Mitchell P. Levesque,Reinhard Dummer,Lukas Flatz
DOI: https://doi.org/10.1186/s40425-019-0523-2
IF: 12.469
2019-02-20
Journal for ImmunoTherapy of Cancer
Abstract:BackgroundLong-term survival of stage IV melanoma patients has improved significantly with the development of immune checkpoint inhibitors (CIs). Reliable biomarkers to predict response and clinical outcome are needed.MethodsWe investigated the role of melanoma-associated antibodies as predictive markers for CI therapy in two independent cohorts. In cohort 1, a prospective study, we measured specific antibodies before treatment, after one week and after six to nine weeks of treatment. Cohort 2 consisted of serum samples prior to CI therapy initiation. ELISA assays were performed to quantify specific IgG directed against melanocyte differentiation antigens tyrosinase-related proteins 1 and 2 (TRP1/TYRP1 and TRP2/TYRP2), glycoprotein 100 (gp100), MelanA/MART1, and the cancer-testis antigen NY-ESO-1. Response was defined as either complete or partial remission on CT scan according to RECIST 1.1.ResultsIn cohort 1, baseline levels of these antibodies were higher in the responder group, although statistical significance was only reached for NY-ESO-1 (p = 0.007). In cohort 2, significantly higher antibody baseline levels for MelanA/MART1 (p = 0.003) and gp100 (p = 0.029) were found. After pooling the results from both cohorts, higher levels of MelanA/MART1 (p = 0.013), TRP1/TYRP1 (p = 0.048), TRP2/TYRP2 (p = 0.047) and NY-ESO-1 (p = 0.005) specific antibodies at baseline were independently associated with response.ConclusionsMelanoma-associated antibodies may be candidate biomarkers for response and survival in metastatic melanoma patients being treated with CIs. These markers may be used to complement patient assessment, in combination with PD-L1 status, tumor-infiltrating lymphocytes and tumor mutational burden, with the aim to predict outcome of CI treatment in patients with metastatic melanoma.Trial registrationEthikkommission Ostschweiz, EKOS 16/079 https://ongoingprojects.swissethics.ch/runningProjects_list.php?q=%28BASECID~contains~2016-00998%29&orderby=dBASECID.
oncology,immunology
-
Perspectives in Melanoma: meeting report from the Melanoma Bridge (December 2nd - 4th, 2021, Italy)
Paolo A Ascierto,Sanjiv S Agarwala,Christian Blank,Corrado Caracò,Richard D Carvajal,Marc S Ernstoff,Soldano Ferrone,Bernard A Fox,Thomas F Gajewski,Claus Garbe,Jean-Jacques Grob,Omid Hamid,Michelle Krogsgaard,Roger S Lo,Amanda W Lund,Gabriele Madonna,Olivier Michielin,Bart Neyns,Iman Osman,Solange Peters,Poulikos I Poulikakos,Sergio A Quezada,Bradley Reinfeld,Laurence Zitvogel,Igor Puzanov,Magdalena Thurin
DOI: https://doi.org/10.1186/s12967-022-03592-4
2022-09-04
Abstract:Advances in immune checkpoint and combination therapy have led to improvement in overall survival for patients with advanced melanoma. Improved understanding of the tumor, tumor microenvironment and tumor immune-evasion mechanisms has resulted in new approaches to targeting and harnessing the host immune response. Combination modalities with other immunotherapy agents, chemotherapy, radiotherapy, electrochemotherapy are also being explored to overcome resistance and to potentiate the immune response. In addition, novel approaches such as adoptive cell therapy, oncogenic viruses, vaccines and different strategies of drug administration including sequential, or combination treatment are being tested. Despite the progress in diagnosis of melanocytic lesions, correct classification of patients, selection of appropriate adjuvant and systemic theràapies, and prediction of response to therapy remain real challenges in melanoma. Improved understanding of the tumor microenvironment, tumor immunity and response to therapy has prompted extensive translational and clinical research in melanoma. There is a growing evidence that genomic and immune features of pre-treatment tumor biopsies may correlate with response in patients with melanoma and other cancers, but they have yet to be fully characterized and implemented clinically. Development of novel biomarker platforms may help to improve diagnostics and predictive accuracy for selection of patients for specific treatment. Overall, the future research efforts in melanoma therapeutics and translational research should focus on several aspects including: (a) developing robust biomarkers to predict efficacy of therapeutic modalities to guide clinical decision-making and optimize treatment regimens, (b) identifying mechanisms of therapeutic resistance to immune checkpoint inhibitors that are potentially actionable, (c) identifying biomarkers to predict therapy-induced adverse events, and (d) studying mechanism of actions of therapeutic agents and developing algorithms to optimize combination treatments. During the Melanoma Bridge meeting (December 2nd-4th, 2021, Naples, Italy) discussions focused on the currently approved systemic and local therapies for advanced melanoma and discussed novel biomarker strategies and advances in precision medicine as well as the impact of COVID-19 pandemic on management of melanoma patients.
-
Early disappearance of tumor antigen-reactive T cells from peripheral blood correlates with superior clinical outcomes in melanoma under anti-PD-1 therapy
Jonas Bochem,Henning Zelba,Janine Spreuer,Teresa Amaral,Nikolaus B Wagner,Andrea Gaissler,Oltin T Pop,Karolin Thiel,Can Yurttas,Daniel Soffel,Stephan Forchhammer,Tobias Sinnberg,Heike Niessner,Friedegund Meier,Patrick Terheyden,Alfred Königsrainer,Claus Garbe,Lukas Flatz,Graham Pawelec,Thomas K Eigentler,Markus W Löffler,Benjamin Weide,Kilian Wistuba-Hamprecht
DOI: https://doi.org/10.1136/jitc-2021-003439
Abstract:Background: Anti-programmed cell death protein 1 (PD-1) antibodies are now routinely administered for metastatic melanoma and for increasing numbers of other cancers, but still only a fraction of patients respond. Better understanding of the modes of action and predictive biomarkers for clinical outcome is urgently required. Cancer rejection is mostly T cell-mediated. We previously showed that the presence of NY-ESO-1-reactive and/or Melan-A-reactive T cells in the blood correlated with prolonged overall survival (OS) of patients with melanoma with a heterogeneous treatment background. Here, we investigated whether such reactive T cells can also be informative for clinical outcomes in metastatic melanoma under PD-1 immune-checkpoint blockade (ICB). Methods: Peripheral blood T cell stimulation by NY-ESO-1 and Melan-A overlapping peptide libraries was assessed before and during ICB in two independent cohorts of a total of 111 patients with stage IV melanoma. In certain cases, tumor-infiltrating lymphocytes could also be assessed for such responses. These were characterized using intracellular cytokine staining for interferon gamma (IFN-γ), tumor negrosis factor (TNF) and CD107a. Digital pathology analysis was performed to quantify NY-ESO-1 and Melan-A expression by tumors. Endpoints were OS and progression-free survival (PFS). Results: The initial presence in the circulation of NY-ESO-1- or Melan-A-reactive T cells which became no longer detectable during ICB correlated with validated, prolonged PFS (HR:0.1; p>0.0001) and OS (HR:0.2; p=0.021). An evaluation of melanoma tissue from selected cases suggested a correlation between tumor-resident NY-ESO-1- and Melan-A-reactive T cells and disease control, supporting the notion of a therapy-associated sequestration of cells from the periphery to the tumor predominantly in those patients benefitting from ICB. Conclusions: Our findings suggest a PD-1 blockade-dependent infiltration of melanoma-reactive T cells from the periphery into the tumor and imply that this seminally contributes to effective treatment.
-
Immunotherapy in melanoma: Can we predict response to treatment with circulating biomarkers?
Elena Splendiani,Zein Mersini Besharat,Alessia Covre,Michele Maio,Anna Maria Di Giacomo,Elisabetta Ferretti
DOI: https://doi.org/10.1016/j.pharmthera.2024.108613
IF: 13.4
2024-02-18
Pharmacology & Therapeutics
Abstract:Melanoma is the most aggressive form of skin cancer, representing approximately 4% of all cutaneous neoplasms it accounts for up to 80% of deaths. Advanced stages of melanoma involve metastatic processes and are associated with high mortality and morbidity, mainly due to the rapid dissemination and heterogeneous responses to current therapies including immunotherapy. Indeed, immune checkpoints inhibitors (ICIs), either alone or in combination with other therapies are currently used in metastatic melanoma (MM) and linked to an increase in patient survival. Of note, the number of therapeutic regimens for MM patients using ICIs has increased, highlighting a growing need for reliable biomarkers that can both predict and monitor response to ICIs. In this context, circulating biomarkers, such as DNA, RNA, proteins, and cells, have emerged due to their ability to detect disease status. Moreover, blood tests are minimally invasive and provide an attractive option to detect biomarkers, avoiding stressful medical procedures. This systematic review summarizes of this review is to evaluate the possibility of a non-invasive biomarker signature that can guide therapeutic decisions. The studies reported here offer valuable insight into how circulating biomarkers can have a role personalized treatments for melanoma patients receiving ICIs therapy, emphasizing the need for rigorous clinical trials to confirm findings and establish standardized procedures.
pharmacology & pharmacy
-
Effectiveness of immune checkpoint inhibitors and other treatment modalities in patients with advanced mucosal melanomas: a systematic review and individual patient data meta-analysis
Andrea York Tiang Teo,Chun En Yau,Chen Ee Low,Jarett Vanz-Brian Pereira,Julia Yu Xin Ng,Tse Kiat Soong,Jack Yu Tung Lo,Valerie Shiwen Yang
DOI: https://doi.org/10.1016/j.eclinm.2024.102870
IF: 15.1
2024-10-04
EClinicalMedicine
Abstract:Background: Mucosal melanomas (MM) are an aggressive subtype of melanoma. Given the rarity of this disease, the conduct of clinical trials is challenging and has been limited. Current treatment options have been extrapolated from the more common cutaneous melanoma even though MM is distinct in pathogenesis, etiology and prognosis. This is the first meta-analysis to comprehensively assess the efficacy of immune checkpoint inhibitors (anti-PD1 and anti-CTLA4) and other treatment modalities (targeted therapy such as KIT inhibitors and VEGF inhibitors, as well as radiotherapy) on survival outcomes in MM to develop clinical guidelines for evidence-based management. Methods: The protocol was prospectively registered on PROSPERO (PROSPERO ID: CRD42023411195). PubMed, Embase, Cochrane Central Register of Controlled Trials (CENTRAL), Web of Science and Google Scholar were searched from inception until 25 July 2024, for all cohort and observational studies. Eligible studies included those with five or more participants with locally advanced or metastatic MM treated with anti-PD1, anti-CTLA4, VEGF inhibitors and/or KIT inhibitors. Titles and abstracts of potential articles were screened and full texts of all potentially eligible studies were retrieved and reviewed by two independent reviewers. Individual patient data (IPD) from published Kaplan-Meier curves were reconstructed using a graphical reconstruction method and pooled as a one-stage meta-analysis. A sensitivity analysis using a two-stage meta-analysis approach was conducted. Extracted outcomes included overall survival (OS) and progression-free survival (PFS). For each treatment arm, median survival time and 12-month survival proportion were estimated. Data from double-arm trials was pooled to estimate hazard ratios (HRs), ratios of restricted mean time lost (RMTL) and restricted mean survival time (RMST). Findings: From a total of 7402 studies, 35 eligible studies comprising a total of 2833 participants were included. Combined anti-PD1 and anti-CTLA4 therapy had the highest 12-month OS and 12-month PFS at 71.8% (95% CI: 67.6%, 76.2%, n = 476) and 35.1% (95% CI: 30.5%, 40.4%, n = 401) respectively, followed by anti-PD1 therapy alone (OS: 64.0% (95% CI: 61.4%, 66.7%, n = 1399); PFS: was 28.3% (95% CI: 25.8%, 31.2%, n = 1142), anti-PD1 and VEGF inhibitor combination therapy (OS: 57.1% (95% CI: 51.0%, 63.9%)), KIT inhibitors (OS: 48.2% (95% CI: 37.6%, 61.8%); PFS: 8.3% (95% CI: 3.7%, 18.7%)) and anti-CTLA4 therapy alone (OS: 33.3% (95% CI: 28.4%, 39.1%); PFS: 9.8% (95% CI: 5.9%, 16.5%)). In the double-arm studies, combination therapy with anti-PD1 and anti-CTLA4 had similar OS and PFS with anti-PD1 alone (OS: HR 0.856 (95% CI: 0.704, 1.04); RMTL ratio 0.932 (95% CI: 0.832, 1.044, P = 0.225); RMST ratio 1.102 (95% CI: 0.948, 1.281, P = 0.204); PFS: HR 0.919 (95% CI: 0.788, 1.07); RMTL ratio 0.936 (95% CI: 0.866, 1.013, P = 0.100); RMST ratio 1.21 (95% CI: 0.979, 1.496, P = 0.078)), however, anti-PD1 therapy alone had significantly better PFS than anti-CTLA4 alone (HR 0.548 (95% CI: 0.376, 0.799); RMTL ratio 0.715 (95% CI: 0.606, 0.844, P < 0.001); RMST ratio 1.659 (95% CI: 1.316, 2.092, P < 0.001)). Anti-PD1 therapy with radiotherapy versus anti-PD1 alone showed no significant difference (OS: HR 0.854 (95% CI: 0.567, 1.29); RMTL ratio 0.855 (95% CI: 0.675, 1.083, P = 0.193); RMST ratio 1.194 (95% CI: 0.928, 1.536, P = 0.168; PFS: HR 0.994 (95% CI: 0.710, 1.39); RMTL ratio 1.006 (95% CI: 0.87, 1.162, P = 0.939); RMST ratio 0.984 (95% CI: 0.658, 1.472, P = 0.939)). Interpretation: For the systemic treatment of MM, anti-PD1 is the best monotherapy. While combining anti-PD1 with other treatment options such as anti-CTLA4, VEGF inhibitors or radiotherapy might achieve better outcomes, these improvements did not reach statistical significance when evaluated by HR, RMTL and RMST ratios. Funding: This work was supported by the National Medical Research Council Transition Award (TA20nov-0020), SingHealth Duke-NUS Oncology Academic Clinical Programme (08/FY2020/EX/67-A143 and 08/FY2021/EX/17-A47), the Khoo Pilot Collaborative Award (Duke-NUS-KP(Coll)/2022/0020A), the National Medical Research Council Clinician Scientist-Individual Research Grant-New Investigator Grant (CNIGnov-0025), the Terry Fox Grant (I1056) and the Khoo Bridge Funding Award (Duke-NUS-KBrFA/2024/0083I).
-
Anti-PD-1 and Novel Combinations in the Treatment of Melanoma—An Update
Frank Gellrich,Marc Schmitz,Stefan Beissert,Friedegund Meier
DOI: https://doi.org/10.3390/jcm9010223
IF: 3.9
2020-01-14
Journal of Clinical Medicine
Abstract:Until recently, distant metastatic melanoma was considered refractory to systemic therapy. A better understanding of the interactions between tumors and the immune system and the mechanisms of regulation of T-cells led to the development of immune checkpoint inhibitors. This review summarizes the current novel data on the treatment of metastatic melanoma with anti-programmed cell death protein 1 (PD-1) antibodies and anti-PD-1-based combination regimens, including clinical trials presented at major conference meetings. Immune checkpoint inhibitors, in particular anti-PD-1 antibodies such as pembrolizumab and nivolumab and the combination of nivolumab with the anti-cytotoxic T-lymphocyte-associated protein 4 (CTLA-4) antibody ipilimumab can achieve long-term survival for patients with metastatic melanoma. The anti-PD-1 antibodies nivolumab and pembrolizumab were also approved for adjuvant treatment of patients with resected metastatic melanoma. Anti-PD-1 antibodies appear to be well tolerated, and toxicity is manageable. Nivolumab combined with ipilimumab achieves a 5 year survival rate of more than 50% but at a cost of high toxicity. Ongoing clinical trials investigate novel immunotherapy combinations and strategies (e.g., Talimogene laherparepvec (T-VEC), Bempegaldesleukin (BEMPEG), incorporation or sequencing of targeted therapy, incorporation or sequencing of radiotherapy), and focus on poor prognosis groups (e.g., high tumor burden/LDH levels, anti-PD-1 refractory melanoma, and brain metastases).
medicine, general & internal
-
Abstract 4527: CD8 T cell-melanoma cell interactions in response and resistance to ipilimumab plus nivolumab: Biopsy analysis of SWOG S1616
Katie M. Campbell,Lawrence Kuklinski,Zaid Bustami,Jessica Maxey,Egmidio Medina,Sandra Santulli-Marotto,Cynthia R. Gonzalez,Nataly Naser Aldeen,Kari Kendra,Sapna Patel,Siwen Hu-Lieskovan,James Moon,Shay Bellasea,Christine N. Spencer,Marshall A. Thompson,Michael Wu,Ari Vanderwalde,Philip O. Scumpia,Antoni Ribas
DOI: https://doi.org/10.1158/1538-7445.am2023-4527
IF: 11.2
2023-04-04
Cancer Research
Abstract:Abstract Background: In the randomized S1616 trial (NCT03033576), patients with metastatic melanoma refractory to anti-PD-1-based therapy had improved progression free survival (PFS, HR = 0.63, p = 0.037) and objective response to ipilimumab plus nivolumab (ipi/nivo, RR 28%) compared to ipilimumab alone (ipi, RR = 9%). We hypothesized that reversal of resistance to PD-1 blockade with CTLA-4 blockade would result in increased CD8 T-cell infiltration in patient biopsies. Methods: Multiplex ion beam imaging (MIBI) for 32 protein markers, spanning tumor, immune, and stromal cell types, was used to evaluate the tumor microenvironment in melanoma tumor biopsies collected at baseline (N=21 samples) and one month on-treatment (N=22; N=16 paired timepoints). Tumor-immune cell dynamics were determined by comparing baseline and on-treatment biopsies within clinical groups (ipi/nivo response, N=10; ipi/nivo no-response, N=9; ipi no-response, N=8). Results: Multiplex analysis in ipi/nivo responsive patients demonstrated increased proportions of CD8 (median 1.6X increase), CD4 (2.1X), regulatory T cells (1.6X), and monocytes (1.4X), with a paralleled 8.1X decrease in melanoma cells over the course of treatment. This was confirmed by histopathologic evidence of tumor regression, necrosis, and immune infiltrate in on-treatment, responding tumor biopsies, as determined by dermatopathologists. Melanoma cells had direct, cell-membrane interactions with CD8 T cells in ipi/nivo responsive patients, both at baseline (median 26%) and on-treatment (median 86%). The shared interface between melanoma and CD8 T-cells had polarized expression of CD3, CD8, and CD45RO. There was no change in T-cell infiltration nor tumor cell content over the course of treatment in ipi/nivo non-responders nor ipi non-responders; similar proportions of melanoma cells were interacting with CD8 T-cells at baseline (median 24%) and on-treatment (27%). Conclusion: Reversal of resistance to PD-1 blockade is associated with increased frequency of CD8 T-melanoma cell interactions and pathological response, reflective of different stages of antitumor immune responses. Response to ipi/nivo in the anti-PD-1 refractory setting was further associated with increased tumor-polarized and activated CD8 T cells interacting with melanoma cells, demonstrated by the colocalized expression of immune synapse proteins at tumor-CD8-T-cell membrane interfaces. Citation Format: Katie M. Campbell, Lawrence Kuklinski, Zaid Bustami, Jessica Maxey, Egmidio Medina, Sandra Santulli-Marotto, Cynthia R. Gonzalez, Nataly Naser Aldeen, Kari Kendra, Sapna Patel, Siwen Hu-Lieskovan, James Moon, Shay Bellasea, Christine N. Spencer, Marshall A. Thompson, Michael Wu, Ari Vanderwalde, Philip O. Scumpia, Antoni Ribas. CD8 T cell-melanoma cell interactions in response and resistance to ipilimumab plus nivolumab: Biopsy analysis of SWOG S1616. [abstract]. In: Proceedings of the American Association for Cancer Research Annual Meeting 2023; Part 1 (Regular and Invited Abstracts); 2023 Apr 14-19; Orlando, FL. Philadelphia (PA): AACR; Cancer Res 2023;83(7_Suppl):Abstract nr 4527.
oncology