Abstract
Precision oncology involves the use of predictive biomarkers to personalize treatment. However, for most cancer therapeutics or combination regimens, effective biomarkers have been elusive. This challenge has fuelled efforts to interrogate increasingly diverse and complex clinical and molecular determinants of treatment response. Some molecular predictors have been identified (for example, based on analysis of transcriptomic or imaging data), although the limited reproducibility and robustness of many of these candidate biomarkers make them difficult to apply in clinical practice. Moreover, different types of predictor must often be combined to optimize treatment selection (for example, gene signatures plus patient characteristics). Computational methods, including machine learning and artificial intelligence approaches, provide opportunities to identify predictive patterns in both clinical data and preclinical datasets and to predict treatment response for individual patients. Such approaches also offer opportunities to predict the efficacy or synergy of drug combinations, for example, via extrapolation from correlations of monotherapy responses or by linking the cellular responses observed in preclinical drug screens with molecular and clinical data from patients. In this Review, we describe the application of computational methods to predictive biomarker discovery, including current progress, key challenges facing this field, and future opportunities.
Key points
-
The discovery and validation of predictive biomarkers is an essential prerequisite to advances in personalized cancer therapies.
-
Predictive biomarkers can be derived from analyses of molecular, radiological and/or histopathological data as well as clinical data, and multiple modalities can be combined to capture more complex biological associations.
-
Both preclinical models and patient data are useful sources for the discovery of predictive biomarkers, and several publicly available collections exist that can also support biomarker investigations.
-
Computational methods and machine learning algorithms can be used to predict responses to treatment both with monotherapies and combination regimens.
-
Several remaining challenges relating to the discovery of predictive biomarkers must be addressed in order to improve the generalizability of predictive models, to better capture tumour heterogeneity and to overcome the various limitations that can hinder clinical translation.
This is a preview of subscription content, access via your institution
Access options
Access Nature and 54 other Nature Portfolio journals
Get Nature+, our best-value online-access subscription
$32.99 / 30 days
cancel any time
Subscribe to this journal
Receive 12 print issues and online access
$189.00 per year
only $15.75 per issue
Buy this article
- Purchase on SpringerLink
- Instant access to the full article PDF.
USD 39.95
Prices may be subject to local taxes which are calculated during checkout



Similar content being viewed by others
References
Ashley, E. A. Towards precision medicine. Nat. Rev. Genet. 17, 507–522 (2016).
Ballman, K. V. Biomarker: predictive or prognostic? J. Clin. Oncol. 33, 3968–3971 (2015).
Drilon, A. et al. Efficacy of larotrectinib in TRK fusion–positive cancers in adults and children. N. Engl. J. Med. 378, 731–739 (2018).
Meric-Bernstam, F. et al. Efficacy and safety of trastuzumab deruxtecan in patients with HER2-expressing solid tumors: primary results from the DESTINY-PanTumor02 phase II trial. J. Clin. Oncol. 42, 47–58 (2024).
Hilas, O. Trends in FDA-approved cancer therapies. U.S. Pharmacist https://www.uspharmacist.com/article/trends-in-fdaapproved-cancer-therapies (2023).
Mullard, A. NCI-MATCH trial pushes cancer umbrella trial paradigm. Nat. Rev. Drug Discov. 14, 513–515 (2015).
Tsimberidou, A.-M. et al. Personalized medicine for patients with advanced cancer in the phase I program at MD Anderson: validation and landmark analyses. Clin. Cancer Res. 20, 4827–4836 (2014).
Stockley, T. L. et al. Molecular profiling of advanced solid tumors and patient outcomes with genotype-matched clinical trials: the Princess Margaret IMPACT/COMPACT trial. Genome Med. 8, 109 (2016).
Weiss, F., Lauffenburger, D. & Friedl, P. Towards targeting of shared mechanisms of cancer metastasis and therapy resistance. Nat. Rev. Cancer 22, 157–173 (2022).
Schmitt, M. W., Loeb, L. A. & Salk, J. J. The influence of subclonal resistance mutations on targeted cancer therapy. Nat. Rev. Clin. Oncol. 13, 335–347 (2016).
De Roock, W., De Vriendt, V., Normanno, N., Ciardiello, F. & Tejpar, S. KRAS, BRAF, PIK3CA, and PTEN mutations: implications for targeted therapies in metastatic colorectal cancer. Lancet Oncol. 12, 594–603 (2011).
Ding, M. Q., Chen, L., Cooper, G. F., Young, J. D. & Lu, X. Precision oncology beyond targeted therapy: combining omics data with machine learning matches the majority of cancer cells to effective therapeutics. Mol. Cancer Res. 16, 269–278 (2018).
Adam, G. et al. Machine learning approaches to drug response prediction: challenges and recent progress. NPJ Precis. Oncol. 4, 19 (2020).
Bhinder, B., Gilvary, C., Madhukar, N. S. & Elemento, O. Artificial intelligence in cancer research and precision medicine. Cancer Discov. 11, 900–915 (2021).
Tran, K. A. et al. Deep learning in cancer diagnosis, prognosis and treatment selection. Genome Med. 13, 152 (2021).
Chiu, Y.-C. et al. Predicting drug response of tumors from integrated genomic profiles by deep neural networks. BMC Med. Genomics 12, 18 (2019).
Kuenzi, B. M. et al. Predicting drug response and synergy using a deep learning model of human cancer cells. Cancer Cell 38, 672–684.e6 (2020).
Alix-Panabières, C. & Pantel, K. Advances in liquid biopsy: from exploration to practical application. Cancer Cell 43, 161–165 (2024).
Passaro, A. et al. Cancer biomarkers: emerging trends and clinical implications for personalized treatment. Cell 187, 1617–1635 (2024).
Mateo, J. et al. Delivering precision oncology to patients with cancer. Nat. Med. 28, 658–665 (2022).
Cai, Z., Poulos, R. C., Liu, J. & Zhong, Q. Machine learning for multi-omics data integration in cancer. iScience 25, 103798 (2022).
Jee, J. et al. Automated real-world data integration improves cancer outcome prediction. Nature 636, 728–736 (2024).
Dumbrava, E. E. I. et al. Targeting ERBB2 (HER2) amplification identified by next-generation sequencing in patients with advanced or metastatic solid tumors beyond conventional indications. JCO Precis. Oncol. 3, PO.18.00345 (2019).
Suehnholz, S. P. et al. Quantifying the expanding landscape of clinical actionability for patients with cancer. Cancer Discov. 14, 49–65 (2024).
Vandereyken, K., Sifrim, A., Thienpont, B. & Voet, T. Methods and applications for single-cell and spatial multi-omics. Nat. Rev. Genet. 24, 494–515 (2023).
Shiao, S. L. et al. Single-cell and spatial profiling identify three response trajectories to pembrolizumab and radiation therapy in triple negative breast cancer. Cancer Cell 42, 70–84.e8 (2024).
Zuo, C. et al. stClinic dissects clinically relevant niches by integrating spatial multi-slice multi-omics data in dynamic graphs. Nat. Commun. 16, 5317 (2025).
Cescon, D. W., Bratman, S. V., Chan, S. M. & Siu, L. L. Circulating tumor DNA and liquid biopsy in oncology. Nat. Cancer 1, 276–290 (2020).
Feng, L. et al. Development and validation of a radiopathomics model to predict pathological complete response to neoadjuvant chemoradiotherapy in locally advanced rectal cancer: a multicentre observational study. Lancet Digit. Health 4, e8–e17 (2022).
Vanguri, R. S. et al. Multimodal integration of radiology, pathology and genomics for prediction of response to PD-(L)1 blockade in patients with non-small cell lung cancer. Nat. Cancer 3, 1151–1164 (2022).
Hwang, C.-I., Boj, S. F., Clevers, H. & Tuveson, D. A. Preclinical models of pancreatic ductal adenocarcinoma. J. Pathol. 238, 197–204 (2016).
Gengenbacher, N., Singhal, M. & Augustin, H. G. Preclinical mouse solid tumour models: status quo, challenges and perspectives. Nat. Rev. Cancer 17, 751–765 (2017).
Mirabelli, P., Coppola, L. & Salvatore, M. Cancer cell lines are useful model systems for medical research. Cancers 11, 1098 (2019).
Domcke, S., Sinha, R., Levine, D. A., Sander, C. & Schultz, N. Evaluating cell lines as tumour models by comparison of genomic profiles. Nat. Commun. 4, 2126 (2013).
Jin, H. et al. Systematic transcriptional analysis of human cell lines for gene expression landscape and tumor representation. Nat. Commun. 14, 5417 (2023).
Mouradov, D. et al. Colorectal cancer cell lines are representative models of the main molecular subtypes of primary cancer. Cancer Res. 74, 3238–3247 (2014).
Najgebauer, H. et al. CELLector: genomics-guided selection of cancer in vitro models. Cell Syst. 10, 424–432.e6 (2020).
Warren, A. et al. Global computational alignment of tumor and cell line transcriptional profiles. Nat. Commun. 12, 22 (2021).
Mourragui, S., Loog, M., van de Wiel, M. A., Reinders, M. J. T. & Wessels, L. F. A. PRECISE: a domain adaptation approach to transfer predictors of drug response from pre-clinical models to tumors. Bioinformatics 35, i510–i519 (2019).
Ma, J. et al. Few-shot learning creates predictive models of drug response that translate from high-throughput screens to individual patients. Nat. Cancer 2, 233–244 (2021).
Ashok, G. & Ramaiah, S. A critical review of datasets and computational suites for improving cancer theranostics and biomarker discovery. Med. Oncol. 39, 206 (2022).
Ghandi, M. et al. Next-generation characterization of the Cancer Cell Line Encyclopedia. Nature 569, 503–508 (2019).
Iorio, F. et al. A landscape of pharmacogenomic interactions in cancer. Cell 166, 740–754 (2016).
Shoemaker, R. H. The NCI60 human tumour cell line anticancer drug screen. Nat. Rev. Cancer 6, 813–823 (2006).
Klijn, C. et al. A comprehensive transcriptional portrait of human cancer cell lines. Nat. Biotechnol. 33, 306–312 (2015).
Dixit, A. et al. Perturb-Seq: dissecting molecular circuits with scalable single-cell RNA profiling of pooled genetic screens. Cell 167, 1853–1866.e17 (2016).
Shalem, O. et al. Genome-scale CRISPR-Cas9 knockout screening in human cells. Science 343, 84–87 (2014).
Funk, J. S. et al. Deep CRISPR mutagenesis characterizes the functional diversity of TP53 mutations. Nat. Genet. 57, 140–153 (2025).
Bottomly, D. et al. Integrative analysis of drug response and clinical outcome in acute myeloid leukemia. Cancer Cell 40, 850–864.e9 (2022).
Pemovska, T. et al. Axitinib effectively inhibits BCR-ABL1(T315I) with a distinct binding conformation. Nature 519, 102–105 (2015).
Lee, S. et al. High-throughput identification of repurposable neuroactive drugs with potent anti-glioblastoma activity. Nat. Med. 30, 3196–3208 (2024).
Acanda De La Rocha, A. M. et al. Feasibility of functional precision medicine for guiding treatment of relapsed or refractory pediatric cancers. Nat. Med. 30, 990–1000 (2024).
Dolgin, E. The future of precision cancer therapy might be to try everything. Nature 626, 470–473 (2024).
Kytölä, S. et al. Ex vivo venetoclax sensitivity predicts clinical response in acute myeloid leukemia in the prospective VenEx trial. Blood 145, 409–421 (2025).
Lee, S. H. R. et al. Pharmacotypes across the genomic landscape of pediatric acute lymphoblastic leukemia and impact on treatment response. Nat. Med. 29, 170–179 (2023).
Sarno, F. et al. A phase III randomized trial of integrated genomics and avatar models for personalized treatment of pancreatic cancer: the AVATAR trial. Clin. Cancer Res. 31, 278–287 (2025).
Ayala Solares, J. R. et al. Deep learning for electronic health records: a comparative review of multiple deep neural architectures. J. Biomed. Inf. 101, 103337 (2020).
Bareche, Y. et al. Leveraging big data of immune checkpoint blockade response identifies novel potential targets. Ann. Oncol. 33, 1304–1317 (2022).
Litchfield, K. et al. Meta-analysis of tumor- and T cell-intrinsic mechanisms of sensitization to checkpoint inhibition. Cell 184, 596–614.e14 (2021).
Snyder, A. et al. Genetic basis for clinical response to CTLA-4 blockade in melanoma. N. Engl. J. Med. 371, 2189–2199 (2014).
Le, D. T. et al. Mismatch repair deficiency predicts response of solid tumors to PD-1 blockade. Science 357, 409–413 (2017).
Hoppe, M. M., Sundar, R., Tan, D. S. P. & Jeyasekharan, A. D. Biomarkers for homologous recombination deficiency in cancer. J. Natl Cancer Inst. 110, 704–713 (2018).
Telli, M. L. et al. Homologous recombination deficiency (HRD) score predicts response to platinum-containing neoadjuvant chemotherapy in patients with triple-negative breast cancer. Clin. Cancer Res. 22, 3764–3773 (2016).
Figueroa, M. E. et al. DNA methylation signatures identify biologically distinct subtypes in acute myeloid leukemia. Cancer Cell 17, 13–27 (2010).
Liberzon, A. et al. The molecular signatures database (MSigDB) hallmark gene set collection. Cell Syst. 1, 417–425 (2015).
Macintyre, G. et al. Copy number signatures and mutational processes in ovarian carcinoma. Nat. Genet. 50, 1262–1270 (2018).
Jiang, P. et al. Signatures of T cell dysfunction and exclusion predict cancer immunotherapy response. Nat. Med. 24, 1550–1558 (2018).
Alexandrov, L. B. et al. Signatures of mutational processes in human cancer. Nature 500, 415–421 (2013).
Alexandrov, L. B. et al. The repertoire of mutational signatures in human cancer. Nature 578, 94–101 (2020).
Billan, S., Kaidar-Person, O. & Gil, Z. Treatment after progression in the era of immunotherapy. Lancet Oncol. 21, e463–e476 (2020).
Borcoman, E. et al. Novel patterns of response under immunotherapy. Ann. Oncol. 30, 385–396 (2019).
Nathan, P. et al. Overall survival benefit with tebentafusp in metastatic uveal melanoma. N. Engl. J. Med. 385, 1196–1206 (2021).
van ’t Veer, L. J. et al. Gene expression profiling predicts clinical outcome of breast cancer. Nature 415, 530–536 (2002).
Cardoso, F. et al. 70-Gene signature as an aid to treatment decisions in early-stage breast cancer. N. Engl. J. Med. 375, 717–729 (2016).
Davies, H. et al. HRDetect is a predictor of BRCA1 and BRCA2 deficiency based on mutational signatures. Nat. Med. 23, 517–525 (2017).
Zhao, E. Y. et al. Homologous recombination deficiency and platinum-based therapy outcomes in advanced breast cancer. Clin. Cancer Res. 23, 7521–7530 (2017).
Patsouris, A. et al. Rucaparib in patients presenting a metastatic breast cancer with homologous recombination deficiency, without germline BRCA1/2 mutation. Eur. J. Cancer 159, 283–295 (2021).
Smirnov, P. et al. Evaluation of statistical approaches for association testing in noisy drug screening data. BMC Bioinformatics 23, 188 (2022).
Anagnostou, V. et al. Multimodal genomic features predict outcome of immune checkpoint blockade in non-small-cell lung cancer. Nat. Cancer 1, 99–111 (2020).
Slamon, D. J. et al. Human breast cancer: correlation of relapse and survival with amplification of the HER-2/neu oncogene. Science 235, 177–182 (1987).
Maemondo, M. et al. Gefitinib or chemotherapy for non-small-cell lung cancer with mutated EGFR. N. Engl. J. Med. 362, 2380–2388 (2010).
Flaherty, K. T. et al. Improved survival with MEK inhibition in BRAF-mutated melanoma. N. Engl. J. Med. 367, 107–114 (2012).
Paik, S. et al. A multigene assay to predict recurrence of tamoxifen-treated, node-negative breast cancer. N. Engl. J. Med. 351, 2817–2826 (2004).
Parker, J. S. et al. Supervised risk predictor of breast cancer based on intrinsic subtypes. J. Clin. Oncol. 27, 1160–1167 (2009).
Filipits, M. et al. A new molecular predictor of distant recurrence in ER-positive, HER2-negative breast cancer adds independent information to conventional clinical risk factors. Clin. Cancer Res. 17, 6012–6020 (2011).
Christopoulos, P. et al. Plasma proteome-based test for first-line treatment selection in metastatic non-small cell lung cancer. JCO Precis. Oncol. 8, e2300555 (2024).
Ali, M. & Aittokallio, T. Machine learning and feature selection for drug response prediction in precision oncology applications. Biophys. Rev. 11, 31–39 (2019).
Greener, J. G., Kandathil, S. M., Moffat, L. & Jones, D. T. A guide to machine learning for biologists. Nat. Rev. Mol. Cell Biol. 23, 40–55 (2022).
Camacho, D. M., Collins, K. M., Powers, R. K., Costello, J. C. & Collins, J. J. Next-generation machine learning for biological networks. Cell 173, 1581–1592 (2018).
Sharifi-Noghabi, H. et al. Drug sensitivity prediction from cell line-based pharmacogenomics data: guidelines for developing machine learning models. Brief. Bioinform. 22, bbab294 (2021).
Ishwaran, H., Kogalur, U. B., Blackstone, E. H. & Lauer, M. S. Random survival forests. Ann. Appl. Stat. 2, 841–860 (2008).
Pölsterl, S., Navab, N. & Katouzian, A. In: Machine Learning and Knowledge Discovery in Databases (eds Appice, A. et al.) 243–259 (Springer, 2015).
Unger, M. & Kather, J. N. A systematic analysis of deep learning in genomics and histopathology for precision oncology. BMC Med. Genomics 17, 48 (2024).
Katzman, J. L. et al. DeepSurv: personalized treatment recommender system using a Cox proportional hazards deep neural network. BMC Med. Res. Methodol. 18, 24 (2018).
Schmauch, B. et al. A deep learning model to predict RNA-Seq expression of tumours from whole slide images. Nat. Commun. 11, 3877 (2020).
Hoang, D.-T. et al. Prediction of DNA methylation-based tumor types from histopathology in central nervous system tumors with deep learning. Nat. Med. 30, 1952–1961 (2024).
Coudray, N. et al. Classification and mutation prediction from non-small cell lung cancer histopathology images using deep learning. Nat. Med. 24, 1559–1567 (2018).
Fu, Y. et al. Pan-cancer computational histopathology reveals mutations, tumor composition and prognosis. Nat. Cancer 1, 800–810 (2020).
Kather, J. N. et al. Pan-cancer image-based detection of clinically actionable genetic alterations. Nat. Cancer 1, 789–799 (2020).
Hoang, D.-T. et al. A deep-learning framework to predict cancer treatment response from histopathology images through imputed transcriptomics. Nat. Cancer 5, 1305–1317 (2024).
Vaswani, A. et al. Attention is all you need. In Proc. 31st Conference on Neural Information Processing Systems 6000–6010 (NeurIPS, 2017).
Yang, X. et al. A large language model for electronic health records. NPJ Digit. Med. 5, 194 (2022).
Pai, S. et al. Foundation model for cancer imaging biomarkers. Nat. Mach. Intell. 6, 354–367 (2024).
Chen, R. J. et al. Towards a general-purpose foundation model for computational pathology. Nat. Med. 30, 850–862 (2024).
Xu, H. et al. A whole-slide foundation model for digital pathology from real-world data. Nature 630, 181–188 (2024).
Wang, X. et al. A pathology foundation model for cancer diagnosis and prognosis prediction. Nature 634, 970–978 (2024).
Cui, H. et al. scGPT: toward building a foundation model for single-cell multi-omics using generative AI. Nat. Methods 21, 1470–1480 (2024).
Theodoris, C. V. et al. Transfer learning enables predictions in network biology. Nature 618, 616–624 (2023).
Hao, M. et al. Large-scale foundation model on single-cell transcriptomics. Nat. Methods 21, 1481–1491 (2024).
Wang, C. et al. ScGPT-spatial: continual pretraining of single-cell foundation model for spatial transcriptomics. Preprint at bioRxiv https://doi.org/10.1101/2025.02.05.636714 (2025).
Moor, M. et al. Foundation models for generalist medical artificial intelligence. Nature 616, 259–265 (2023).
van de Haar, J. et al. Combining genomic biomarkers to guide immunotherapy in non-small cell lung cancer. Clin. Cancer Res. 30, 1307–1318 (2024).
Usset, J. et al. Five latent factors underlie response to immunotherapy. Nat. Genet. 56, 2112–2120 (2024).
Chang, T.-G. et al. LORIS robustly predicts patient outcomes with immune checkpoint blockade therapy using common clinical, pathologic and genomic features. Nat. Cancer 5, 1158–1175 (2024).
Steyaert, S. et al. Multimodal data fusion for cancer biomarker discovery with deep learning. Nat. Mach. Intell. 5, 351–362 (2023).
Lipkova, J. et al. Artificial intelligence for multimodal data integration in oncology. Cancer Cell 40, 1095–1110 (2022).
Carrillo-Perez, F. et al. Machine-learning-based late fusion on multi-omics and multi-scale data for non-small-cell lung cancer diagnosis. J. Pers. Med. 12, 601 (2022).
Cheerla, A. & Gevaert, O. Deep learning with multimodal representation for pancancer prognosis prediction. Bioinformatics 35, i446–i454 (2019).
Ma, S. et al. Moving towards genome-wide data integration for patient stratification with Integrate Any Omics. Nat. Mach. Intell. 7, 29–42 (2025).
Xu, P., Zhu, X. & Clifton, D. A. Multimodal learning with Transformers: a survey. IEEE Trans. Pattern Anal. Mach. Intell. 45, 12113–12132 (2023).
Xiang, J. et al. A vision-language foundation model for precision oncology. Nature 638, 769–778 (2025).
Cui, H. et al. Towards multimodal foundation models in molecular cell biology. Nature 640, 623–633 (2025).
Arango-Argoty, G. et al. Pretrained transformers applied to clinical studies improve predictions of treatment efficacy and associated biomarkers. Nat. Commun. 16, 2101 (2025).
Boehm, K. M., Khosravi, P., Vanguri, R., Gao, J. & Shah, S. P. Harnessing multimodal data integration to advance precision oncology. Nat. Rev. Cancer 22, 114–126 (2022).
Singhal, A. et al. The hallmarks of predictive oncology. Cancer Discov. 15, 271–285 (2025).
Pudjihartono, N., Fadason, T., Kempa-Liehr, A. W. & O’Sullivan, J. M. A review of feature selection methods for machine learning-based disease risk prediction. Front. Bioinform. 2, 927312 (2022).
Cao, Y., Geddes, T. A., Yang, J. Y. H. & Yang, P. Ensemble deep learning in bioinformatics. Nat. Mach. Intell. 2, 500–508 (2020).
Sammut, S.-J. et al. Multi-omic machine learning predictor of breast cancer therapy response. Nature 601, 623–629 (2022).
Zhang, Y. & Yang, Q. An overview of multi-task learning. Natl Sci. Rev. 5, 30–43 (2018).
Thung, K.-H. & Wee, C.-Y. A brief review on multi-task learning. Multimed. Tools Appl. 77, 29705–29725 (2018).
White, B. S. et al. Bayesian multi-source regression and monocyte-associated gene expression predict BCL-2 inhibitor resistance in acute myeloid leukemia. NPJ Precis. Oncol. 5, 71 (2021).
Costello, J. C. et al. A community effort to assess and improve drug sensitivity prediction algorithms. Nat. Biotechnol. 32, 1202–1212 (2014).
Kowald, A. et al. Transfer learning of clinical outcomes from preclinical molecular data, principles and perspectives. Brief. Bioinform. 23, bbac133 (2022).
Kirkpatrick, J. et al. Overcoming catastrophic forgetting in neural networks. Proc. Natl Acad. Sci. USA 114, 3521–3526 (2017).
Li, Z. & Hoiem, D. Learning without forgetting. IEEE Trans. Pattern Anal. Mach. Intell. 40, 2935–2947 (2018).
Rolnick, D., Ahuja, A., Schwarz, J., Lillicrap, T. P. & Wayne, G. Experience replay for continual learning. In Proc. 33rd Conference on Neural Information Processing Systems 350–360 (NeurIPS 2019).
Rusu, A. A. et al. Progressive neural networks. Preprint at https://doi.org/10.48550/arXiv.1606.04671 (2016).
Lopez-Paz, D. & Ranzato, M. Gradient episodic memory for continual learning. In Proc. 31st International Conference on Neural Information Processing Systems 6470–6479 (NeurIPS, 2017).
Xu, Y. et al. Deep learning predicts lung cancer treatment response from serial medical imaging. Clin. Cancer Res. 25, 3266–3275 (2019).
Kummar, S. et al. Utilizing targeted cancer therapeutic agents in combination: novel approaches and urgent requirements. Nat. Rev. Drug Discov. 9, 843–856 (2010).
Jin, H., Wang, L. & Bernards, R. Rational combinations of targeted cancer therapies: background, advances and challenges. Nat. Rev. Drug Discov. 22, 213–234 (2023).
Conroy, T. et al. FOLFIRINOX or gemcitabine as adjuvant therapy for pancreatic cancer. N. Engl. J. Med. 379, 2395–2406 (2018).
Von Hoff, D. D. et al. Increased survival in pancreatic cancer with nab-paclitaxel plus gemcitabine. N. Engl. J. Med. 369, 1691–1703 (2013).
Schmid, P. et al. Pembrolizumab for early triple-negative breast cancer. N. Engl. J. Med. 382, 810–821 (2020).
Antonia, S. J. et al. Overall survival with durvalumab after chemoradiotherapy in stage III NSCLC. N. Engl. J. Med. 379, 2342–2350 (2018).
Loupakis, F. et al. Initial therapy with FOLFOXIRI and bevacizumab for metastatic colorectal cancer. N. Engl. J. Med. 371, 1609–1618 (2014).
Jaaks, P. et al. Effective drug combinations in breast, colon and pancreatic cancer cells. Nature 603, 166–173 (2022).
Sicklick, J. K. et al. Molecular profiling of advanced malignancies guides first-line N-of-1 treatments in the I-PREDICT treatment-naïve study. Genome Med. 13, 155 (2021).
Sicklick, J. K. et al. Molecular profiling of cancer patients enables personalized combination therapy: the I-PREDICT study. Nat. Med. 25, 744–750 (2019).
Meric-Bernstam, F. et al. National Cancer Institute combination therapy platform trial with molecular analysis for therapy choice (ComboMATCH). Clin. Cancer Res. 29, 1412–1422 (2023).
Li, X. et al. Precision combination therapies based on recurrent oncogenic coalterations. Cancer Discov. 12, 1542–1559 (2022).
US National Library of Medicine. ClinicalTrials.gov https://clinicaltrials.gov/study/NCT05674825 (2025).
Menden, M. P. et al. Community assessment to advance computational prediction of cancer drug combinations in a pharmacogenomic screen. Nat. Commun. 10, 2674 (2019).
Kong, W. et al. Systematic review of computational methods for drug combination prediction. Comput. Struct. Biotechnol. J. 20, 2807–2814 (2022).
Dinstag, G. et al. Clinically oriented prediction of patient response to targeted and immunotherapies from the tumor transcriptome. Med 4, 15–30.e8 (2023).
Kuru, H. I., Cicek, A. E. & Tastan, O. From cell lines to cancer patients: personalized drug synergy prediction. Bioinformatics 40, btae134 (2022).
Chen, Y. et al. A machine learning-based strategy predicts selective and synergistic drug combinations for relapsed acute myeloid leukemia. Cancer Res. 85, 2753–2768 (2025).
Pomeroy, A. E., Schmidt, E. V., Sorger, P. K. & Palmer, A. C. Drug independence and the curability of cancer by combination chemotherapy. Trends Cancer 8, 915–929 (2022).
Bunne, C. et al. Learning single-cell perturbation responses using neural optimal transport. Nat. Methods 20, 1759–1768 (2023).
Roohani, Y., Huang, K. & Leskovec, J. Predicting transcriptional outcomes of novel multigene perturbations with GEARS. Nat. Biotechnol. 42, 927–935 (2024).
Zhang, W. et al. Integration of pan-cancer cell line and single-cell transcriptomic profiles enables inference of therapeutic vulnerabilities in heterogeneous tumors. Cancer Res. 84, 2021–2033 (2024).
Schrod, S., Zacharias, H. U., Beißbarth, T., Hauschild, A.-C. & Altenbuchinger, M. CODEX: counterfactual deep learning for the in silico exploration of cancer cell line perturbations. Bioinformatics 40, i91–i99 (2024).
Maeser, D., Zhang, W., Huang, Y. & Huang, R. S. A review of computational methods for predicting cancer drug response at the single-cell level through integration with bulk RNAseq data. Curr. Opin. Struct. Biol. 84, 102745 (2024).
Ianevski, A. et al. Patient-tailored design for selective co-inhibition of leukemic cell subpopulations. Sci. Adv. 7, eabe4038 (2021).
Sinha, S. et al. PERCEPTION predicts patient response and resistance to treatment using single-cell transcriptomics of their tumors. Nat. Cancer 5, 938–952 (2024).
Ianevski, A. et al. Single-cell transcriptomes identify patient-tailored therapies for selective co-inhibition of cancer clones. Nat. Commun. 15, 8579 (2024).
Kornauth, C. et al. Functional precision medicine provides clinical benefit in advanced aggressive hematologic cancers and identifies exceptional responders. Cancer Discov. 12, 372–387 (2022).
Miglino, N. et al. Feasibility of multiomics tumor profiling for guiding treatment of melanoma. Nat. Med. 31, 2430–2441 (2025).
Sheridan, C. Can single-cell biology realize the promise of precision medicine? Nat. Biotechnol. 42, 159–162 (2024).
PromethION. Oxford Nanopore Technologies https://nanoporetech.com/products/sequence/promethion (2025).
Tang, C. et al. Personalized tumor combination therapy optimization using the single-cell transcriptome. Genome Med. 15, 105 (2023).
Tatarova, Z. et al. A multiplex implantable microdevice assay identifies synergistic combinations of cancer immunotherapies and conventional drugs. Nat. Biotechnol. 40, 1823–1833 (2022).
Lundberg, S. & Lee, S.-I. A unified approach to interpreting model predictions. In Proc. 31st International Conference on Neural Information Processing Systems 4765–4774 (2017).
Ribeiro, M. T., Singh, S. & Guestrin, C. Why should I trust you?’: explaining the predictions of any classifier. In Proc. 22nd ACM SIGKDD International Conference on Knowledge Discovery and Data Mining. Vol. 16, 1135–1144 (Association for Computing Machinery, 2016).
Zhou, B., Khosla, A., Lapedriza, A., Oliva, A. & Torralba, A. Learning deep features for discriminative localization. In Proc. IEEE Conference on Computer Vision and Pattern Recognition 2921–2929 (IEEE, 2016).
Tomaszewski, M. R. & Gillies, R. J. The biological meaning of radiomic features. Radiology 298, 505–516 (2021).
Ancona, M., Ceolini, E., Öztireli, C. & Gross, M. Towards better understanding of gradient-based attribution methods for deep neural networks. in Proc. 6th International Conference on Learning Representations https://doi.org/10.3929/ethz-b-000249929 (ICLR, 2018).
Shrikumar, A., Greenside, P. & Kundaje, A. Learning important features through propagating activation differences. In Proc. 34th International Conference on Machine Learning 3145–3153 (PMLR, 2017).
Elmarakeby, H. A. et al. Biologically informed deep neural network for prostate cancer discovery. Nature 598, 348–352 (2021).
Park, S. et al. A deep learning model of tumor cell architecture elucidates response and resistance to CDK4/6 inhibitors. Nat. Cancer 5, 996–1009 (2024).
Haibe-Kains, B. et al. Transparency and reproducibility in artificial intelligence. Nature 586, E14–E16 (2020).
Wilkinson, M. D. et al. The FAIR guiding principles for scientific data management and stewardship. Sci. Data 3, 160018 (2016).
Rieke, N. et al. The future of digital health with federated learning. NPJ Digit. Med. 3, 119 (2020).
Ogier du Terrail, J. et al. Federated learning for predicting histological response to neoadjuvant chemotherapy in triple-negative breast cancer. Nat. Med. 29, 135–146 (2023).
Terry Fox Research Institute Marathon of Hope Cancer Centers Network. The Terry Fox Research Institute Marathon of Hope Cancer Centres Network: a pan-Canadian precision oncology initiative. Cancer Cell 43, 587–592 (2025).
Mahon, P. et al. A federated learning system for precision oncology in Europe: DigiONE. Nat. Med. 30, 334–337 (2024).
Wen, J. et al. A survey on federated learning: challenges and applications. Int. J. Mach. Learn. Cybern. 14, 513–535 (2023).
Moon, I. et al. Machine learning for genetics-based classification and treatment response prediction in cancer of unknown primary. Nat. Med. 29, 2057–2067 (2023).
Ianni, J. D. et al. Tailored for real-world: a whole slide image classification system validated on uncurated multi-site data emulating the prospective pathology workload. Sci. Rep. 10, 3217 (2020).
Feuerriegel, S. et al. Causal machine learning for predicting treatment outcomes. Nat. Med. 30, 958–968 (2024).
Lone, S. N. et al. Liquid biopsy: a step closer to transform diagnosis, prognosis and future of cancer treatments. Mol. Cancer 21, 79 (2022).
Sanz-Garcia, E., Zhao, E., Bratman, S. V. & Siu, L. L. Monitoring and adapting cancer treatment using circulating tumor DNA kinetics: current research, opportunities, and challenges. Sci. Adv. 8, eabi8618 (2022).
Rozenblatt-Rosen, O. et al. The human tumor atlas network: charting tumor transitions across space and time at single-cell resolution. Cell 181, 236–249 (2020).
Frampton, G. M. et al. Development and validation of a clinical cancer genomic profiling test based on massively parallel DNA sequencing. Nat. Biotechnol. 31, 1023–1031 (2013).
Malone, E. R., Oliva, M., Sabatini, P. J. B., Stockley, T. L. & Siu, L. L. Molecular profiling for precision cancer therapies. Genome Med. 12, 8 (2020).
Cai, Z. et al. Synthetic augmentation of cancer cell line multi-omic datasets using unsupervised deep learning. Nat. Commun. 15, 10390 (2024).
Thangaraj, P. M., Shankar, S. V., Oikonomou, E. K. & Khera, R. RCT-Twin-GAN generates digital twins of randomized control trials adapted to real-world patients to enhance their inference and application. Preprint at medRxiv https://doi.org/10.1101/2023.12.06.23299464 (2023).
AACR Project GENIE Consortium. AACR Project GENIE: powering precision medicine through an international consortium. Cancer Discov. 7, 818–831 (2017).
Turnbull, C. et al. The 100 000 genomes project: bringing whole genome sequencing to the NHS. BMJ 361, k1687 (2018).
Topol, E. J. Welcoming new guidelines for AI clinical research. Nat. Med. 26, 1318–1320 (2020).
Liu, X. et al. Reporting guidelines for clinical trial reports for interventions involving artificial intelligence: the CONSORT-AI extension. Nat. Med. 26, 1364–1374 (2020).
Cruz Rivera, S. et al. Guidelines for clinical trial protocols for interventions involving artificial intelligence: the SPIRIT-AI extension. Nat. Med. 26, 1351–1363 (2020).
Collins, G. S. et al. TRIPOD+AI statement: updated guidance for reporting clinical prediction models that use regression or machine learning methods. BMJ 385, e078378 (2024).
Gallifant, J. et al. The TRIPOD-LLM reporting guideline for studies using large language models. Nat. Med. 31, 60–69 (2025).
Garassino, M. C. et al. PL02.11 Normalized membrane ratio of TROP2 by quantitative continuous scoring is predictive of clinical outcomes in TROPION-lung 01. J. Thorac. Oncol. 19, S2–S3 (2024).
Foy, R., Lew, K. X. & Saurin, A. T. The search for CDK4/6 inhibitor biomarkers has been hampered by inappropriate proliferation assays. NPJ Breast Cancer 10, 19 (2024).
U.S. Food and Drug Administration. Principles for Codevelopment of an In Vitro Companion Diagnostic Device with a Therapeutic Product https://www.fda.gov/regulatory-information/search-fda-guidance-documents/principles-codevelopment-in-vitro-companion-diagnostic-device-therapeutic-product (2019).
Maliepaard, M., Nibi, P., Nibi, G. & Pasmooij, A. M. G. Evaluation of companion diagnostics in scientific advice and drug marketing authorization applications by the European Medicines Agency. Front. Med. 9, 893028 (2022).
Horgan, D. et al. Bringing greater accuracy to Europe’s healthcare systems: the unexploited potential of biomarker testing in oncology. Biomed. Hub. 5, 182–223 (2020).
de Moor, J. S., Gray, S. W., Mitchell, S. A., Klabunde, C. N. & Freedman, A. N. Oncologist confidence in genomic testing and implications for using multimarker tumor panel tests in practice. JCO Precis. Oncol. 4, 620–631 (2020).
Mateo, J. et al. A framework to rank genomic alterations as targets for cancer precision medicine: the ESMO Scale for Clinical Actionability of molecular Targets (ESCAT). Ann. Oncol. 29, 1895–1902 (2018).
Chakravarty, D. et al. OncoKB: a precision oncology knowledge base. JCO Precis. Oncol. https://doi.org/10.1200/PO.17.00011 (2017).
Asghar, U. S. & Chung, C. Application of digital twins for personalized oncology. Nat. Rev. Cancer 25, 823–825 (2025).
Bhatt, D. L. & Mehta, C. Adaptive designs for clinical trials. N. Engl. J. Med. 375, 65–74 (2016).
Thorlund, K., Haggstrom, J., Park, J. J. & Mills, E. J. Key design considerations for adaptive clinical trials: a primer for clinicians. BMJ 360, k698 (2018).
Wang, H. & Yee, D. I-SPY 2: a neoadjuvant adaptive clinical trial designed to improve outcomes in high-risk breast cancer. Curr. Breast Cancer Rep. 11, 303–310 (2019).
Marret, G., Herrera, M. & Siu, L. L. Turning the kaleidoscope: innovations shaping the future of clinical trial design. Cancer Cell 43, 597–605 (2025).
Drukker, C. A. et al. A prospective evaluation of a breast cancer prognosis signature in the observational RASTER study. Int. J. Cancer 133, 929–936 (2013).
Sparano, J. A. et al. Prospective validation of a 21-gene expression assay in breast cancer. N. Engl. J. Med. 373, 2005–2014 (2015).
Alvarez, M. J. et al. A precision oncology approach to the pharmacological targeting of mechanistic dependencies in neuroendocrine tumors. Nat. Genet. 50, 979–989 (2018).
Kather, J. N. et al. Deep learning can predict microsatellite instability directly from histology in gastrointestinal cancer. Nat. Med. 25, 1054–1056 (2019).
Mu, W. et al. Non-invasive measurement of PD-L1 status and prediction of immunotherapy response using deep learning of PET/CT images. J. Immunother. Cancer 9, e002118 (2021).
Wagner, S. J. et al. Transformer-based biomarker prediction from colorectal cancer histology: a large-scale multicentric study. Cancer Cell 41, 1650–1661.e4 (2023).
Sun, R. et al. A radiomics approach to assess tumour-infiltrating CD8 cells and response to anti-PD-1 or anti-PD-L1 immunotherapy: an imaging biomarker, retrospective multicohort study. Lancet Oncol. 19, 1180–1191 (2018).
Preuer, K. et al. DeepSynergy: predicting anti-cancer drug synergy with deep learning. Bioinformatics 34, 1538–1546 (2018).
Xia, F. et al. Predicting tumor cell line response to drug pairs with deep learning. BMC Bioinformatics 19, 486 (2018).
Zhang, T., Zhang, L., Payne, P. R. O. & Li, F. Synergistic drug combination prediction by integrating multiomics data in deep learning models. Methods Mol. Biol. 2194, 223–238 (2021).
Kim, Y. et al. Anticancer drug synergy prediction in understudied tissues using transfer learning. J. Am. Med. Inf. Assoc. 28, 42–51 (2021).
Yang, J., Xu, Z., Wu, W. K. K., Chu, Q. & Zhang, Q. GraphSynergy: a network-inspired deep learning model for anticancer drug combination prediction. J. Am. Med. Inf. Assoc. 28, 2336–2345 (2021).
Liu, Q. & Xie, L. TranSynergy: mechanism-driven interpretable deep neural network for the synergistic prediction and pathway deconvolution of drug combinations. PLoS Comput. Biol. 17, e1008653 (2021).
Kuru, H. I., Tastan, O. & Cicek, A. E. MatchMaker: a deep learning framework for drug synergy prediction. IEEE/ACM Trans. Comput. Biol. Bioinform. 19, 2334–2344 (2022).
Wang, X. et al. PRODeepSyn: predicting anticancer synergistic drug combinations by embedding cell lines with protein-protein interaction network. Brief. Bioinform. 23, bbab587 (2022).
Hu, J. et al. DTSyn: a dual-transformer-based neural network to predict synergistic drug combinations. Brief. Bioinform. 23, bbac302 (2022).
Tang, Y.-C. & Gottlieb, A. SynPathy: predicting drug synergy through drug-associated pathways using deep learning. Mol. Cancer Res. 20, 762–769 (2022).
Hosseini, S.-R. & Zhou, X. CCSynergy: an integrative deep-learning framework enabling context-aware prediction of anti-cancer drug synergy. Brief. Bioinform. 24, bbac588 (2023).
Xu, M. et al. DFFNDDS: prediction of synergistic drug combinations with dual feature fusion networks. J. Cheminform. 15, 33 (2023).
El Khili, M. R., Memon, S. A. & Emad, A. MARSY: a multitask deep-learning framework for prediction of drug combination synergy scores. Bioinformatics 39, btad177 (2023).
Zhang, P. & Tu, S. MGAE-DC: predicting the synergistic effects of drug combinations through multi-channel graph autoencoders. PLoS Comput. Biol. 19, e1010951 (2023).
Li, T.-H., Wang, C.-C., Zhang, L. & Chen, X. SNRMPACDC: computational model focused on Siamese network and random matrix projection for anticancer synergistic drug combination prediction. Brief. Bioinform. 24, bbac503 (2023).
Verkerk, K. & Voest, E. E. Generating and using real-world data: a worthwhile uphill battle. Cell 187, 1636–1650 (2024).
Yu, Y. et al. Association of survival and immune-related biomarkers with immunotherapy in patients with non-small cell lung cancer: a meta-analysis and individual patient-level analysis. JAMA Netw. Open 2, e196879 (2019).
Acknowledgements
We sincerely thank L. Siu of the Princess Margaret Cancer Centre for her insightful comments on this Review. The authors’ research is supported by funding from the ARPA-H ADAPT Program (OT 140D042590013 to T.I. and B.H.-K.).
Author information
Authors and Affiliations
Contributions
X.W. and J.N. initiated and coordinated the Review. X.W., J.N., K.N., M.M., P.W., A.S., T.A. and B.H.-K. researched data for the manuscript and made a substantial contribution to discussions of content. X.W. and P.L.B. provided clinical input. T.I., T.A. and B.H.-K. supervised the Review. All authors edited and/or reviewed the manuscript prior to submission.
Corresponding authors
Ethics declarations
Competing interests
P.L.B. reports research funding (to the institution) from Amgen, AstraZeneca, Bayer, Boehringer Ingelheim, Bicara Therapeutics, Bristol Myers Squibb, Daiichi Sankyo, Genentech/Roche, GlaxoSmithKline, LegoChem, Lilly, Merck, Medicenna, Novartis, Seagen/Pfizer, Takeda and Zymeworks. He also reports uncompensated honoraria/consultancy with Amgen, Daiichi Sankyo, Gilead, Janssen, Lilly, Merck, Repare, Seattle Genetics and Zymeworks, and a compensated advisory role for Boehringer Ingelheim. T.I. is a co-founder, member of the advisory board, and has an equity interest in Data4Cure and Serinus Biosciences, is a consultant of and has an equity interest in Ideaya Biosciences, and is a member of the advisory editorial board of The EMBO Journal. The terms of all consultancy and equity arrangements have been reviewed and approved by the University of California, San Diego in accordance with its conflict-of-interest policies. B.H.-K. is part of the scientific advisory boards of Break Through Cancer, Commonwealth Cancer Consortium (USA), Consortium de Recherche Biopharmaceutique (CQDM; Quebec, Canada), the Canadian Institute of Health Research — Institute of Genetics, Cancer Grand Challenges (UK) and Chriners Children (Florida, USA), is a co-founder of the MAQC (Massive Analysis and Quality Control) Society and is part of the board of directors of AACR International (Canada) and The American Association for Cancer Research (USA). X.W., J.N., K.N., M.M., P.W., A.S. and T.A. declare no competing interests.
Peer review
Peer review information
Nature Reviews Clinical Oncology thanks C. Cheng, V. Fortino, J. Kather and the other, anonymous, reviewer(s) for their contribution to the peer review of this work.
Additional information
Publisher’s note Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.
Rights and permissions
Springer Nature or its licensor (e.g. a society or other partner) holds exclusive rights to this article under a publishing agreement with the author(s) or other rightsholder(s); author self-archiving of the accepted manuscript version of this article is solely governed by the terms of such publishing agreement and applicable law.
About this article
Cite this article
Wang, X., Nguyen, J., Nader, K. et al. Discovery of predictive biomarkers for cancer therapy through computational approaches. Nat Rev Clin Oncol (2026). https://doi.org/10.1038/s41571-025-01109-8
Accepted:
Published:
Version of record:
DOI: https://doi.org/10.1038/s41571-025-01109-8


