Cancer is not an isolated collection of mutated cells. It exists and evolves within a highly dynamic and interactive ecosystem known as the tumor microenvironment (TME). The TME surrounds and sustains tumors, playing a central role in cancer occurrence, progression, invasion, metastasis, and even therapeutic resistance [1-4].
How does the TME drive cancer progression? And why is this understanding crucial for developing more effective cancer treatments?
Table of Contents
1. What Is Tumor Microenvironment?
2. Mechanisms by Which the Tumor Microenvironment Drives Cancer Progression
3. Heterogeneity of the Tumor Microenvironment
The tumor microenvironment (TME) is a complex and dynamic ecosystem that surrounds and interacts with tumor cells within the body. It mainly includes cellular and noncellular components, including immune cells, fibroblasts, extracellular matrix (ECM), blood vessels, and soluble factors. The TME evolves continuously through crosstalk between all these components, influencing the tumor's behavior and survival [7].
Figure 1. Important components of TME and their interaction with tumor cells
The components of the TME may vary by tissue type and co-evolve as the tumor progresses. The various cells in the TME can be tumor-suppressive or tumor-supportive.
Table 1. Cellular and noncellular components of the TME (Table information is cited from: https://pmc.ncbi.nlm.nih.gov/articles/PMC7199555/)
Category | Components | Function | Description | |
---|---|---|---|---|
Cellular Elements | Tumor cells | The primary culprits, which mutate, invade, and manipulate their surroundings to support their own survival and propagation [5] | Different types | |
Immune cells | Neutrophils | Enhancement of angiogenesis and metastasis; associated with poor prognosis. | Tumor promoting or inhibiting; increased levels in the colon, stomach, and lung cancer patients. | |
Tumor-associated macrophages (TAMs) | Promoting degradation of the extracellular matrix [6,7]; aiding the expansion of inflammatory cytokines, such as TNF-β; enhancement of angiogenesis and remodeling. | The major protumoral component in TME; the first nonneoplastic cells infiltrating the tumor; attracted by chemokines secreted by both malignant and stromal cells. | ||
CD8+ cytotoxic T cells (CTL) | Induce apoptosis, necrosis, and growth arrest by releasing INF-γ and other cytotoxic cytokines; establishing an antitumor environment. | Tumor inhibiting; the major antitumoral component in TME. | ||
Regulatory T cells (Tregs) | Secreting cytokines such as IL-10, TGF-β; establishing an immunosuppressive environment; associated with poor prognosis. | Tumor promoting; promoting tumor maintenance. | ||
Myeloid-derived suppressor cells (MDSCs) | Associated with tumor progression and neoangiogenesis; suppressing T cells and NK cells; differentiating into TAMs under hypoxic conditions. | Tumor promoting; increased in almost all patients/animals with cancer; including premature granulocytes, macrophages, dendritic cells, and myeloid precursors. | ||
Stromal Cells | Cancer-associated fibroblasts (CAFs) [8] | Sustaining proliferative signaling; activating angiogenesis and metastasis; tumor-promoting inflammation; evading immune destruction; reprogramming cellular metabolism; promoting genome instability and mutation. | Pro or anti-tumor activities; commonly used markers including α-, MYL9, MYLK, MMP2, FAP, COL1A2, and PDGFRβ; CAFs are the primary source for the synthesis, secretion, assembly, and modification of the composition and organization of the ECM. | |
Other Cells | Mesenchymal stem cells (MSCs) | Differentiating into mesenchymal tissues such as bone, cartilage, and fat tissues, vasculogenic mimicry; forming the premetastatic niche; promoting cancer initiation and malignancy. | Tumor promoting; the major component of stromal cells in TME. | |
Endothelial cells | Consisting of tumor blood vessels [9]; secreting angiocrine factors such as adhesion molecules; intercommunicating with tumor cells via secreting EVs including CD106, CD49a. | Tumor promoting | ||
Adipocytes | Regulating the balance of systematic energy and metabolism; releasing free fatty acids, cytokines, adipokines, chemokines, growth factors, and hormones; promoting cancer progression. | Tumor promoting. | ||
Neuroendocrine cells (NE cells) | Promoting proliferative signaling; secreting neurotransmitters, including CgA, chromophilic and vasoactive polypeptide; regulating NK cell migration and toxicity ability. | Tumor promoting. | ||
Noncellular Elements | Vascular network | Providing oxygen, clearing carbon dioxide, and metabolizing wastes; providing nutrition support for cancer cells; promoting angiogenesis and metastasis. | Tumor promoting; all malignant tumors are angiogenesis-dependent. | |
Lymph vessels | Helping immune cell avoid immunity and dissemination; providing a physical link between lymph nodes and tumor. | Tumor promoting. | ||
Extracellular vesicles (EVs) | Membrane-wrapped vesicles including exosomes, microvesicles, and apoptotic bodies | Carrying biologically active molecules such as proteins, miRNAs, and lncRNAs from donor cell to recipient cell; regulating key signaling pathways, proliferation, drug resistance, and stemness; reprogramming stromal cells to create a niche for survival. | Tumor promoting or inhibiting; as a critical mediator between tumor and the TME. | |
Extracellular matrix (ECM) | Fibrous proteins (collagen, elastin), glycosaminoglycans (hyaluronic acid), proteoglycans (chondroitin sulfate, heparan sulfate), and glycoproteins (fibronectin 1 (FN1), laminins, tenascin C (TNC)) | Forming the complex macromolecular network; controlling cancer invasion and metastasis, angiogenesis; contribution to growth and proliferation signaling, inhibiting cancer apoptosis [10]. | Tumor promoting. | |
Metabolites | Lactate, glucose, fatty acids, and other metabolic products | Shaping the local environment, acidify tissues, supporting tumor metabolism, and suppressing anti-tumor immunity [11,12]. |
Tumor cells instruct neighboring fibroblasts and immune cells, which modify the ECM and vascular architecture, creating an environment that supports every stage of cancer development [5,7,13]. This synergy forms a living, ever-changing ecosystem where cancer flourishes but also, potentially, can be targeted for destruction.
The TME is a multifaceted and dynamic entity that drives cancer progression through different mechanisms, including immune escape, angiogenesis, metabolic reprogramming, and ECM remodeling.
Normal immune surveillance can destroy emerging cancer cells, but in the TME, immune escape is the rule rather than the exception [14]. Tumors evade destruction by recruiting immunosuppressive populations (like Tregs and M2-type macrophages), secreting cytokines such as IL-10 and TGF-β, and expressing surface proteins (e.g., PD-L1) that inhibit cytotoxic T cell activity [14,15]. These actions shut down immune attack and allow cancer to thrive invisibly [16].
Other mechanisms include exclusion of effector T cells from the tumor mass, exhaustion of cytotoxic T cells, and editing of antigenicity so that tumor cells are no longer recognized as foreign [15].
By deciphering this immune crosstalk, researchers and clinicians can devise therapies like PD-1/PD-L1 inhibitors that reinvigorate T cell activity and overcome immune suppression, improving patient response to immunotherapy [17,18].
As tumors grow, they quickly outstrip their oxygen and nutrient supply [19]. The TME responds by releasing angiogenic factors, notably VEGF, that signal endothelial cells to sprout new blood vessels [20]. These vessels are often irregular, leaky, and dysfunctional, but are essential for feeding the tumor and providing escape routes for metastasizing cells [19,21]. Angiogenesis is a defining hallmark of cancer progression [22,23].
Targeting angiogenic pathways with anti-VEGF drugs (like bevacizumab) can “starve” the tumor, slow progression, and sensitize it to other therapies [24,25].
Cancer cells within the TME undergo metabolic reprogramming to support unchecked growth [26]. This means increased glycolysis (“Warburg effect”), enhanced glutamine and fatty acid uptake, and secretion of acidifying metabolites like lactate [26]. These changes deplete local nutrients, acidify the tissue, and directly suppress immune cell function while promoting invasion [12,27]. Surrounding stromal and immune cells are also metabolically reprogrammed, further buffering tumor growth [28].
Therapies that inhibit glycolysis, glutaminolysis, or fatty acid metabolism, either alone or in combination, can make tumors more vulnerable to immunotherapy and conventional treatments [29-31].The ECM is not inert. In cancer, it is persistently shaped by stromal cells and tumor-derived enzymes [32,33]. CAFs secrete matrix proteins and matrix metalloproteinases (MMPs), both strengthening and degrading the ECM [7]. This remodeling paves the way for tumor cell migration, vascular invasion (intravasation), and eventual metastasis [32]. ECM stiffening and altered signaling through integrins can further induce a more aggressive tumor phenotype.
Understanding ECM dynamics suggests therapeutic avenues like inhibitors of MMPs, signals that block CAF activation, or agents that reverse ECM stiffness, which may limit metastasis and improve therapies [7,33].
TMEs are notoriously heterogeneous [34]. This heterogeneity manifests:
- Across cancer types: Each has a distinct TME "signature," dictated by tissue origin, genetic mutations, and stromal context [35].
- Among patients: Host genetics, lifestyle, microbiome, and co-morbidities create individualized TMEs, affecting therapy response [34].
- Within a single tumor: Different regions may be "hot" (rich in immune cells) or "cold," hypoxic or well-vascularized, fibrotic or loose.
This variability underlies why some patients respond to immunotherapy or chemotherapy while others develop resistance or relapse [36]. Tumor cell clones can dynamically reprogram their microenvironment to exclude drugs, resist immune attack, or promote their survival [37]. As a result, overcoming TME heterogeneity is a major challenge and opportunity for precision oncology [34,38].
The TME has emerged as a critical focus in cancer research and therapy because of its central role in cancer progression, therapeutic resistance, and immune evasion. By targeting the TME, researchers aim to disrupt the supportive ecosystem that tumors rely on, thereby enhancing the effectiveness of existing treatments and overcoming resistance.
Modern cancer immunotherapy often works by releasing the “brakes” imposed by the TME [18,39]. Checkpoint inhibitors targeting PD-1/PD-L1 or CTLA-4 restore T cell recognition and destruction of tumor cells [17,18,40]. The TME's immunosuppressive mechanisms are thus at the very heart of breakthrough cancer treatments [41,42].
By analyzing the TME in each patient, clinicians and researchers can select those who will most benefit from immunotherapy, avoid unnecessary toxicity, and guide combination approaches to overcome resistance [41].
Drugs that block VEGF or related pathways normalize blood vessels, reduce oxygen and nutrient flow to the tumor, and sometimes restore immune cell access [21,25,43]. These therapies not only slow tumor growth but can synergize with immune checkpoint blockade [25].
Combining anti-angiogenic therapy with immunotherapy or chemotherapy is a rational strategy for harder-to-treat tumors [25].
Blocking key metabolic dependencies (such as glycolysis, glutamine metabolism, or lipid synthesis) rewires the TME and makes cancer less able to tolerate additional stress (like immune attack or chemotherapy) [29-31].
Personalized metabolic targeting, ideally guided by TME profiling, could offer new hope to patients resistant to conventional regimens [44,45].
In the future, TME-targeting therapy will involve combinations that are purposefully designed rather than relying on a single-drug approach [42,46].
- Immunotherapy plus anti-angiogenic drugs, to both unleash the immune system and improve its access to the tumor [47].
- Immunotherapy plus metabolic therapy, to weaken both the cancer cells and their suppressive microenvironment [46,47].
- Immune checkpoint blockade plus agents that modulate the ECM or target CAFs, to overcome physical barriers to immune infiltration [47].
However, combination approaches require careful balance, since increased toxicity, complex resistance mechanisms, and dynamic adaptation of the TME remain significant hurdles [42].
The TME's ability to remodel itself means that it is not, in fact, a static foe. Targeting one aspect (fibrosis, angiogenesis, metabolism) can potentiate later attacks, making it an attractive therapeutic target if approached with strategic vision [48].
Despite stunning progress, there remain significant limitations in our understanding. For example, many laboratory models fail to capture the full complexity and dynamic, three-dimensional structure of human TMEs [49,50]. The temporal evolution of the TME—how it adapts and changes during disease progression or treatment—remains poorly understood [49].
Cutting-edge technologies are beginning to address these gaps.
- Single-cell sequencing enables detailed mapping of every cell type within a tumor and its microenvironment, revealing rare populations, novel targets, and resistance mechanisms [51].
- Spatial transcriptomics preserves tissue architecture, allowing researchers to see not just who is present, but where and how different regions (e.g., hypoxic vs. well-oxygenated) interact [52-54].
- Multi-omics integration (combining genomics, transcriptomics, proteomics, and metabolomics) generates comprehensive maps of the molecular state of the TME, guiding biomarker discovery, and the rational design of combinatorial treatments [55-57].
Personalizing cancer treatment based on TME characteristics is the next frontier. By profiling patients, TMEs in detail, clinicians can select the optimal combination of drugs for each individual. This approach promises better outcomes, fewer side effects, and, importantly, new hope for historically "untreatable" cancers.
The TME is not just the backdrop for cancer but is a central driver of its progression, metastasis, and resistance to therapy. Understanding the TME helps researchers develop new, more effective therapies that target cancer’s support system, not just the cancer cells themselves. Clinicians can better personalize treatment, overcoming resistance and reducing unnecessary toxicity. For patients, this means more options, greater hope, and, ultimately, better outcomes.
With advances in multi-omics, spatial transcriptomics, computational biology, and immunology, targeting the TME may offer a once elusive promise—a future where cancer is controlled, chronic, or even cured.
Further Reading:
The Metabolic Phenotype of Tumor-Associated Macrophages in the Tumor Microenvironment
How Does Cytokines Work in Cancer Cells?
What Are Immune Checkpoint Inhibitors?
PD-1-- An Important Immune Checkpoint
PD-1/PD-L1, What a Powerful Immune Checkpoint
References
[1] Avci, C. B., Bagca, et al. (2024). Tumor microenvironment and cancer metastasis: Molecular mechanisms and therapeutic implications [J]. Frontiers in Pharmacology, 15, 1442888.
[2] Wang, Q., Shao, X., et al. (2023). Role of tumor microenvironment in cancer progression and therapeutic strategy [J]. Cancer Medicine, 12(10), 11149.
[3] Li, Z., Li, J., Bai, X. et al. Tumor microenvironment as a complex milieu driving cancer progression: a mini review [J]. Clin Transl Oncol 27, 1943–1952 (2025).
[4] Wang, Q., Shao, X., et al. (2023). Role of tumor microenvironment in cancer progression and therapeutic strategy [J]. Cancer Medicine, 12(10), 11149.
[5] Baghy, K., Ladányi, A., Reszegi, A., & Kovalszky, I. (2022). Insights into the Tumor Microenvironment—Components, Functions and Therapeutics [J]. International Journal of Molecular Sciences, 24(24), 17536.
[6] Yang, Y., Yang, F., Huang, Z., et al. (2023). T cells, NK cells, and tumor-associated macrophages in cancer immunotherapy and the current state of the art of drug delivery systems [J]. Frontiers in Immunology, 14, 1199173.
[7] Anderson, N. M., & Simon, M. C. (2020). Tumor Microenvironment [J]. Current Biology : CB, 30(16), R921.
[8] Sarkar, M., Nguyen, T., et al. (2023). Cancer-associated fibroblasts: The chief architect in the tumor microenvironment [J]. Frontiers in Cell and Developmental Biology, 11, 1089068.
[9] Leone, P., Malerba, E., et al. (2024). Endothelial cells in tumor microenvironment: Insights and perspectives [J]. Frontiers in Immunology, 15, 1367875.
[10] Popova, N. V., & Jücker, M. (2022). The Functional Role of Extracellular Matrix Proteins in Cancer [J]. Cancers, 14(1), 238.
[11] Cortellino, S., & Longo, V. D. (2023). Metabolites and Immune Response in Tumor Microenvironments [J]. Cancers, 15(15), 3898.
[12] C., J., & Lahoz, A. (2020). Tumor Microenvironment-Derived Metabolites: A Guide to Find New Metabolic Therapeutic Targets and Biomarkers [J]. Cancers, 13(13), 3230.
[13] Zhao, Y., Shen, M., et al. (2023). Stromal cells in the tumor microenvironment: Accomplices of tumor progression [J]? Cell Death & Disease, 14(9), 1-24.
[14] Zhou, C., Liu, Q., Xiang, Y., Gou, X., & Li, W. (2021). Role of the tumor immune microenvironment in tumor immunotherapy [J]. Oncology Letters, 23(2), 53.
[15] Spranger, S. (2016). Mechanisms of tumor escape in the context of the T-cell-inflamed and the non-T-cell-inflamed tumor microenvironment [J]. International Immunology, 28(8), 383.
[16] Tang, S., Ning, Q., Yang, L., Mo, Z., & Tang, S. (2020). Mechanisms of immune escape in the cancer immune cycle [J]. International Immunopharmacology, 86, 106700.
[17] Liu, J., Chen, Z., Li, Y., Zhao, W., Wu, J., & Zhang, Z. (2021). PD-1/PD-L1 Checkpoint Inhibitors in Tumor Immunotherapy [J]. Frontiers in Pharmacology, 12, 731798.
[18] Arafat Hossain, M. (2024). A comprehensive review of immune checkpoint inhibitors for cancer treatment [J]. International Immunopharmacology, 143, 113365.
[19] Mahaki, H., Nobari, S., et al. (2025). Targeting VEGF signaling for tumor microenvironment remodeling and metastasis inhibition: Therapeutic strategies and insights [J]. Biomedicine & Pharmacotherapy, 186, 118023.
[20] Schaaf, M. B., Garg, A. D., & Agostinis, P. (2018). Defining the role of the tumor vasculature in antitumor immunity and immunotherapy [J]. Cell Death & Disease, 9(2), 1-14.
[21] Zhang, R., Yao, Y., Gao, H., & Hu, X. (2024). Mechanisms of angiogenesis in tumor [J]. Frontiers in Oncology, 14, 1359069.
[22] Jiang, X., Wang, J., et al. (2020). The role of microenvironment in tumor angiogenesis [J]. Journal of Experimental & Clinical Cancer Research : CR, 39, 204.
[23] Watnick, R. S. (2012). The Role of the Tumor Microenvironment in Regulating Angiogenesis [J]. Cold Spring Harbor Perspectives in Medicine, 2(12), a006676.
[24] Lopes-Coelho, F., Martins, F., Pereira, S. A., & Serpa, J. (2021). Anti-Angiogenic Therapy: Current Challenges and Future Perspectives [J]. International Journal of Molecular Sciences, 22(7), 3765.
[25] Xu, J. (2024). Progress on angiogenic and antiangiogenic agents in the tumor microenvironment [J]. Frontiers in Oncology, 14, 1491099.
[26] Elia, I., & Haigis, M. C. (2020). Metabolites and the tumour microenvironment: From cellular mechanisms to systemic metabolism [J]. Nature Metabolism, 3(1), 21-32.
[27] Arner, E. N., & Rathmell, J. C. (2023). Metabolic programming and immune suppression in the tumor microenvironment [J]. Cancer Cell, 41(3), 421-433.
[28] Wang, J., He, Y., et al. (2024). Metabolic Reprogramming of Immune Cells in the Tumor Microenvironment [J]. International Journal of Molecular Sciences, 25(22), 12223.
[29] Jia, S., Bode, A. M., Chen, X., & Luo, X. (2024). Unlocking the potential: Targeting metabolic pathways in the tumor microenvironment for Cancer therapy [J]. Biochimica et Biophysica Acta (BBA) - Reviews on Cancer, 1879(5), 189166.
[30] El-Tanani, M., Rabbani, S. A., El-Tanani, Y., & Matalka, I. I. (2024). Metabolic vulnerabilities in cancer: A new therapeutic strategy [J]. Critical Reviews in Oncology/Hematology, 201, 104438.
[31] Hyroššová, P., Milošević, M., et al. (2022). Effects of metabolic cancer therapy on tumor microenvironment [J]. Frontiers in Oncology, 12, 1046630.
[32] Di Martino, J. S., Akhter, T., & Bravo-Cordero, J. J. (2021). Remodeling the ECM: Implications for Metastasis and Tumor Dormancy [J]. Cancers, 13(19), 4916.
[33] Kai, F., Drain, A. P., & Weaver, V. M. (2019). The Extracellular Matrix Modulates the Metastatic Journey [J]. Developmental Cell, 49(3), 332-346.
[34] Hass, R., & Ungefroren, H. (2020). Impact of the Tumor Microenvironment on Tumor Heterogeneity and Consequences for Cancer Cell Plasticity and Stemness [J]. Cancers, 12(12), 3716.
[35] Proietto, M., Crippa, M., et al. (2023). Tumor heterogeneity: Preclinical models, emerging technologies, and future applications [J]. Frontiers in Oncology, 13, 1164535.
[36] Maleki, E. H., Bahrami, A. R., & Matin, M. M. (2023). Cancer cell cycle heterogeneity as a critical determinant of therapeutic resistance [J]. Genes & Diseases, 11(1), 189-204.
[37] Vo, J. N., Wu, Y., et al. (2022). The genetic heterogeneity and drug resistance mechanisms of relapsed refractory multiple myeloma [J]. Nature Communications, 13(1), 1-13.
[38] Ge, R., Wang, Z., & Cheng, L. (2022). Tumor microenvironment heterogeneity an important mediator of prostate cancer progression and therapeutic resistance [J]. Npj Precision Oncology, 6(1), 1-8.
[39] R., Y., Warner, A. B., & Wolchok, J. D. (2020). The future of cancer immunotherapy: Microenvironment-targeting combinations [J]. Cell Research, 30(6), 507-519.
[40] Tang, T., Huang, X., et al. (2021). Advantages of targeting the tumor immune microenvironment over blocking immune checkpoint in cancer immunotherapy [J]. Signal Transduction and Targeted Therapy, 6(1), 1-13.
[41] Ye, W., Li, M., & Luo, K. (2023). Therapies Targeting Immune Cells in Tumor Microenvironment for Non-Small Cell Lung Cancer [J]. Pharmaceutics, 15(7), 1788.
[42] Liu, B., Zhou, H., Tan, L., Siu, K. T., & Guan, X. (2024). Exploring treatment options in cancer: Tumor treatment strategies [J]. Signal Transduction and Targeted Therapy, 9(1), 1-44.
[43] Liu, Z., Chen, H., Zheng, L., Sun, L., & Shi, L. (2023). Angiogenic signaling pathways and anti-angiogenic therapy for cancer [J]. Signal Transduction and Targeted Therapy, 8(1), 1-39.
[44] Luo, Z., Eichinger, K. M., Zhang, A., & Li, S. (2023). Targeting cancer metabolic pathways for improving chemotherapy and immunotherapy [J]. Cancer Letters, 575, 216396.
[45] Salemme, V., Centonze, G., et al. (2023). The role of tumor microenvironment in drug resistance: Emerging technologies to unravel breast cancer heterogeneity [J]. Frontiers in Oncology, 13, 1170264.
[46] Roma-Rodrigues, C., Mendes, R., Baptista, P. V., & Fernandes, A. R. (2019). Targeting Tumor Microenvironment for Cancer Therapy [J]. International Journal of Molecular Sciences, 20(4), 840.
[47] Chyuan, T., Chu, L., & Hsu, N. (2021). Targeting the Tumor Microenvironment for Improving Therapeutic Effectiveness in Cancer Immunotherapy: Focusing on Immune Checkpoint Inhibitors and Combination Therapies [J]. Cancers, 13(6), 1188.
[48] Xiao, Y., & Yu, D. (2020). Tumor microenvironment as a therapeutic target in cancer [J]. Pharmacology & Therapeutics, 221, 107753.
[49] Crouigneau, R., Li, Y., et al. (2024). Mimicking and analyzing the tumor microenvironment [J]. Cell Reports Methods, 4(10), 100866.
[50] Ahmed, M. A., & Nagelkerke, A. (2021). Current developments in modelling the tumour microenvironment in vitro: Incorporation of biochemical and physical gradients. Organs-On-A-Chip, 3, 100012.
[51] Chen, S., Zhou, Z., Li, Y., Du, Y., & Chen, G. (2023). Application of single-cell sequencing to the research of tumor microenvironment [J]. Frontiers in Immunology, 14, 1285540.
[52] Zuo, C., Xia, J., & Chen, L. (2024). Dissecting tumor microenvironment from spatially resolved transcriptomics data by heterogeneous graph learning [J]. Nature Communications, 15(1), 1-18.
[53] Yu, Q., Jiang, M., & Wu, L. (2022). Spatial transcriptomics technology in cancer research [J]. Frontiers in Oncology, 12, 1019111.
[54] Huang, S., Ouyang, L., Tang, J. et al. Spatial transcriptomics: a new frontier in cancer research [J]. CCB 3, 13 (2024).
[55] Finotello, F., & Eduati, F. (2018). Multi-Omics Profiling of the Tumor Microenvironment: Paving the Way to Precision Immuno-Oncology. Frontiers in Oncology, 8, 413388.
[56] Sabit, H., Arneth, B., et al. (2024). Leveraging Single-Cell Multi-Omics to Decode Tumor Microenvironment Diversity and Therapeutic Resistance [J]. Pharmaceuticals, 18(1), 75. Fraunhoffer, N. A., Abuelafia, A. M., et al. (2022). Multi-omics data integration and modeling unravels new mechanisms for pancreatic cancer and improves prognostic prediction [J]. Npj Precision Oncology, 6(1), 1-16.
Comments
Leave a Comment