Genomic and Transcriptomic Predictors of Response to Immune Checkpoint Inhibitors in Melanoma Patients: A Machine Learning Approach

被引:6
作者
Ahmed, Yaman B. [1 ]
Al-Bzour, Ayah N. [1 ]
Ababneh, Obada E. [1 ]
Abushukair, Hassan M. [1 ]
Saeed, Anwaar [2 ,3 ]
机构
[1] Jordan Univ Sci & Technol, Fac Med, Irbid 22110, Jordan
[2] Kansas Univ, Canc Ctr, Dept Med, Div Med Oncol, Kansas City, KS 66205 USA
[3] Univ Pittsburgh, UPMC Hillman Canc Ctr, Dept Med, Div Hematol & Oncol, Pittsburgh, PA 15213 USA
关键词
melanoma; immune checkpoint inhibitors; machine learning; tumor mutational burden; TUMOR MUTATIONAL BURDEN; PD-L1; EXPRESSION; CTLA-4; BLOCKADE; BIOMARKER; GPI-80; IMMUNOTHERAPY;
D O I
10.3390/cancers14225605
中图分类号
R73 [肿瘤学];
学科分类号
100214 ;
摘要
Simple Summary Our work provides novel transcriptomic biomarkers that can accurately predict immune checkpoint inhibitors (ICIs) response in melanoma patients. Using a bioinformatics analysis and supervised machine learning approach, we developed four random-forest classifiers based on clinical, genomic, transcriptomic and survival data. The results of these models can enable further insight into the potential role of these genes in immunotherapy. In addition, our findings were based on a supervised approach, in which melanoma patients treated with ICI were used to retrieve response-associated biomarkers, unlike several studies that used an unsupervised approach based on drug targets to predict ICI response in non-ICI-treated melanoma patients. Apart from ICI response, we also investigated the effect of these biomarkers on overall survival and patients' prognosis, which also revealed a high association with survival, marking these biomarkers as powerful in both ICI response and patients' prognosis. Thus, our work demonstrates a cornerstone in precision oncology and further evaluates these biomarkers in clinical practice using personalized medicine for a better prognosis and response outcomes. Immune checkpoint inhibitors (ICIs) became one of the most revolutionary cancer treatments, especially in melanoma. While they have been proven to prolong survival with lesser side effects compared to chemotherapy, the accurate prediction of response remains to be an unmet gap. Thus, we aim to identify accurate clinical and transcriptomic biomarkers for ICI response in melanoma. We also provide mechanistic insight into how high-performing markers impose their effect on the tumor microenvironment (TME). Clinical and transcriptomic data were retrieved from melanoma studies administering ICIs from cBioportal and GEO databases. Four machine learning models were developed using random-forest classification (RFC) entailing clinical and genomic features (RFC7), differentially expressed genes (DEGs, RFC-Seq), survival-related DEGs (RFC-Surv) and a combination model. The xCELL algorithm was used to investigate the TME. A total of 212 ICI-treated melanoma patients were identified. All models achieved a high area under the curve (AUC) and bootstrap estimate (RFC7: 0.71, 0.74; RFC-Seq: 0.87, 0.75; RFC-Surv: 0.76, 0.76, respectively). Tumor mutation burden, GSTA3, and VNN2 were the highest contributing features. Tumor infiltration analyses revealed a direct correlation between upregulated genes and CD8+, CD4+ T cells, and B cells and inversely correlated with myeloid-derived suppressor cells. Our findings confirmed the accuracy of several genomic, clinical, and transcriptomic-based RFC models, that could further support the use of TMB in predicting response to ICIs. Novel genes (GSTA3 and VNN2) were identified through RFC-seq and RFC-surv models that could serve as genomic biomarkers after robust validation.
引用
收藏
页数:20
相关论文
共 50 条
  • [1] [Anonymous], 3 1 CROSS VALIDATION
  • [2] xCell: digitally portraying the tissue cellular heterogeneity landscape
    Aran, Dvir
    Hu, Zicheng
    Butte, Atul J.
    [J]. GENOME BIOLOGY, 2017, 18
  • [3] Understanding the tumor immune microenvironment (TIME) for effective therapy
    Binnewies, Mikhail
    Roberts, Edward W.
    Kersten, Kelly
    Chan, Vincent
    Fearon, Douglas F.
    Merad, Miriam
    Coussens, Lisa M.
    Gabrilovich, Dmitry I.
    Ostrand-Rosenberg, Suzanne
    Hedrick, Catherine C.
    Vonderheide, Robert H.
    Pittet, Mikael J.
    Jain, Rakesh K.
    Zou, Weiping
    Howcroft, T. Kevin
    Woodhouse, Elisa C.
    Weinberg, Robert A.
    Krummel, Matthew F.
    [J]. NATURE MEDICINE, 2018, 24 (05) : 541 - 550
  • [4] MelanA-negative spindle-cell associated melanoma, a distinct inflammatory phenotype correlated with dense infiltration of CD163 macrophages and loss of E-cadherin
    Bonnelykke-Behrndtz, Marie L.
    Steiniche, Torben
    Damsgaard, Tine E.
    Georgsen, Jeanette B.
    Danielsen, Allan
    Bastholt, Lars
    Moller, Holger J.
    Norgaard, Peter H.
    Schmidt, Henrik
    [J]. MELANOMA RESEARCH, 2015, 25 (02) : 113 - 118
  • [5] The hematopoietic stem cell marker VNN2 is associated with chemoresistance in pediatric B-cell precursor ALL
    Bornhauser, Beat
    Cario, Gunnar
    Rinaldi, Anna
    Risch, Thomas
    Martinez, Virginia Rodriguez
    Schutte, Moritz
    Warnatz, Hans-Joerg
    Scheidegger, Nastassja
    Mirkowska, Paulina
    Temperli, Martina
    Moeller, Claudia
    Schumich, Angela
    Dworzak, Michael
    Attarbaschi, Andishe
    Brueggemann, Monika
    Ritgen, Mathias
    Mejstrikova, Ester
    Hofmann, Andreas
    Buldini, Barbara
    Scarparo, Pamela
    Basso, Giuseppe
    Maglia, Oscar
    Gaipa, Giuseppe
    Skoblyn, Tessa-Lara
    te Kronnie, Geertruij
    Vendramini, Elena
    Panzer-Gruemayer, Renate
    Barz, Malwine Jeanette
    Marovca, Blerim
    Hauri-Hohl, Mathias
    Niggli, Felix
    Eckert, Cornelia
    Schrappe, Martin
    Stanulla, Martin
    Zimmermann, Martin
    Wollscheid, Bernd
    Yaspo, Marie-Laure
    Bourquin, Jean-Pierre
    [J]. BLOOD ADVANCES, 2020, 4 (17) : 4052 - 4064
  • [6] Tertiary lymphoid structures improve immunotherapy and survival in melanoma
    Cabrita, Rita
    Lauss, Martin
    Sanna, Adriana
    Donia, Marco
    Larsen, Mathilde Skaarup
    Mitra, Shamik
    Johansson, Iva
    Phung, Bengt
    Harbst, Katja
    Vallon-Christersson, Johan
    van Schoiack, Alison
    Loevgren, Kristina
    Warren, Sarah
    Jirstroem, Karin
    Olsson, Hakan
    Pietras, Kristian
    Ingvar, Christian
    Isaksson, Karolin
    Schadendorf, Dirk
    Schmidt, Henrik
    Bastholt, Lars
    Carneiro, Ana
    Wargo, Jennifer A.
    Svane, Inge Marie
    Jonsson, Goran
    [J]. NATURE, 2020, 577 (7791) : 561 - +
  • [7] Comprehensive Analysis of Hypermutation in Human Cancer
    Campbell, Brittany B.
    Light, Nicholas
    Fabrizio, David
    Zatzman, Matthew
    Fuligni, Fabio
    de Borja, Richard
    Davidson, Scott
    Edwards, Melissa
    Elvin, Julia A.
    Hodel, Karl P.
    Zahurancik, Walter J.
    Suo, Zucai
    Lipman, Tatiana
    Wimmer, Katharina
    Kratz, Christian P.
    Bowers, Daniel C.
    Laetsch, Theodore W.
    Dunn, Gavin P.
    Johanns, Tanner M.
    Grimmer, Matthew R.
    Smirnov, Ivan V.
    Larouche, Valerie
    Samuel, David
    Bronsema, Annika
    Osborn, Michael
    Stearns, Duncan
    Raman, Pichai
    Cole, Kristina A.
    Storm, Phillip B.
    Yalon, Michal
    Opocher, Enrico
    Mason, Gary
    Thomas, Gregory A.
    Sabel, Magnus
    George, Ben
    Ziegler, David S.
    Lindhorst, Scott
    Issai, Vanan Magimairajan
    Constantini, Shlomi
    Toledano, Helen
    Elhasid, Ronit
    Farah, Roula
    Dvir, Rina
    Dirks, Peter
    Huang, Annie
    Galati, Melissa A.
    Chung, Jiil
    Ramaswamy, Vijay
    Irwin, Meredith S.
    Aronson, Melyssa
    [J]. CELL, 2017, 171 (05) : 1042 - +
  • [8] The cBio Cancer Genomics Portal: An Open Platform for Exploring Multidimensional Cancer Genomics Data
    Cerami, Ethan
    Gao, Jianjiong
    Dogrusoz, Ugur
    Gross, Benjamin E.
    Sumer, Selcuk Onur
    Aksoy, Buelent Arman
    Jacobsen, Anders
    Byrne, Caitlin J.
    Heuer, Michael L.
    Larsson, Erik
    Antipin, Yevgeniy
    Reva, Boris
    Goldberg, Arthur P.
    Sander, Chris
    Schultz, Nikolaus
    [J]. CANCER DISCOVERY, 2012, 2 (05) : 401 - 404
  • [9] Chakraborty G, 2020, J CLIN ONCOL, V38
  • [10] Analysis of 100,000 human cancer genomes reveals the landscape of tumor mutational burden
    Chalmers, Zachary R.
    Connelly, Caitlin F.
    Fabrizio, David
    Gay, Laurie
    Ali, Siraj M.
    Ennis, Riley
    Schrock, Alexa
    Campbell, Brittany
    Shlien, Adam
    Chmielecki, Juliann
    Huang, Franklin
    He, Yuting
    Sun, James
    Tabori, Uri
    Kennedy, Mark
    Lieber, Daniel S.
    Roels, Steven
    White, Jared
    Otto, Geoffrey A.
    Ross, Jeffrey S.
    Garraway, Levi
    Miller, Vincent A.
    Stephens, Phillip J.
    Frampton, Garrett M.
    [J]. GENOME MEDICINE, 2017, 9