Key Points on the Hallmarks of Cancer from a Molecular Perspective
- Martin Döhring
- vor 2 Stunden
- 5 Min. Lesezeit

- Research suggests that cancer arises through the acquisition of distinct biological capabilities, often driven by molecular alterations like genetic mutations, epigenetic changes, and signaling pathway disruptions.
- The core hallmarks include sustaining proliferation, evading suppression, resisting death, achieving immortality, inducing angiogenesis, and enabling invasion/metastasis, with emerging ones like metabolic reprogramming and immune evasion.
- Recent updates propose additional dimensions such as phenotypic plasticity, nonmutational epigenetic reprogramming, polymorphic microbiomes, and senescent cells, reflecting ongoing debates about cancer's molecular complexity.
- Evidence leans toward these hallmarks being enabled by genome instability and inflammation, which generate molecular diversity and support tumor progression.
- While widely accepted, the framework acknowledges variability across cancer types, encouraging a balanced view that integrates counterarguments from studies questioning universal applicability.
### Overview of Core Hallmarks
The hallmarks of cancer provide a framework for understanding how normal cells transform into malignant ones at the molecular level. Originally outlined in 2000 and updated in 2011, these capabilities highlight disruptions in cellular regulation. For instance, mutations in genes like RAS or PI3K can sustain proliferative signaling, while inactivation of p53 allows cells to evade growth suppressors. This molecular lens helps explain why cancers resist treatments and spread.
### Emerging and Enabling Features
More recent insights from 2022 suggest additions like unlocking phenotypic plasticity, where epigenetic regulators alter cell states without mutations, potentially enabling adaptability in tumors. Polymorphic microbiomes and senescent cells in the tumor microenvironment also play roles, influencing molecular interactions that foster cancer progression. These elements underscore the interplay between genetics, environment, and cellular behavior.
### Implications for Understanding Cancer
It seems likely that focusing on molecular mechanisms can guide targeted therapies, such as inhibitors for angiogenic factors like VEGF or immune checkpoint blockers. However, the complexity invites consideration of diverse viewpoints, including how non-genetic factors contribute to hallmarks.
---
The hallmarks of cancer represent a foundational framework in oncology, distilling the intricate molecular processes that enable neoplastic transformation and progression. First conceptualized in a seminal 2000 review by Douglas Hanahan and Robert A. Weinberg, the model proposed six acquired capabilities that cancer cells develop during tumorigenesis. This was expanded in 2011 to include two emerging hallmarks and enabling characteristics, reflecting advances in molecular biology. A further refinement in 2022 introduced new dimensions, emphasizing phenotypic plasticity, epigenetic reprogramming, microbiomes, and senescent cells. From a molecular viewpoint, these hallmarks involve genetic mutations, epigenetic modifications, aberrant signaling pathways, and interactions with the tumor microenvironment, providing a lens to rationalize cancer's diversity.
At the core, sustaining proliferative signaling allows cancer cells to deregulate growth controls typically mediated by extracellular signals. Molecularly, this often involves oncogenic activation of receptors (e.g., EGFR) or downstream effectors like RAS and PI3K/AKT pathways, leading to unchecked cell division. Counterbalancing this, evading growth suppressors entails inactivating tumor suppressor genes such as TP53 or RB1, which normally halt proliferation through cell cycle checkpoints involving cyclin-dependent kinases. Resisting cell death, or apoptosis evasion, is achieved via upregulation of anti-apoptotic proteins like BCL-2 or downregulation of pro-apoptotic factors such as BAX, often through PI3K/AKT or NF-κB signaling.
Enabling replicative immortality addresses the telomere crisis, where cancer cells activate telomerase (hTERT) or alternative lengthening mechanisms to maintain chromosome ends, preventing senescence. Inducing angiogenesis involves molecular cues like hypoxia-inducible factor-1α (HIF-1α) upregulating vascular endothelial growth factor (VEGF), promoting new vessel formation to supply nutrients. Activating invasion and metastasis, a critical hallmark, encompasses epithelial-mesenchymal transition (EMT) driven by transcription factors such as SNAIL, TWIST, and ZEB1, which repress E-cadherin and enhance matrix metalloproteinases (MMPs) for extracellular matrix degradation. Molecularly, this process includes chromosomal instability activating NF-κB, exosome-mediated priming of premetastatic niches, and organ-specific tropism via integrins and chemokines.
The 2011 update added reprogramming of energy metabolism, exemplified by the Warburg effect, where cancer cells favor aerobic glycolysis through increased expression of GLUT1 transporters and enzymes like PKM2, supporting biomass production under hypoxic conditions. Evading immune destruction involves molecular strategies like PD-L1 expression to inhibit T-cell activity or secretion of immunosuppressive cytokines such as TGF-β. Enabling these are genome instability (e.g., defects in DNA repair genes like BRCA1/2 leading to mutations) and tumor-promoting inflammation (via cytokines like TNF-α and IL-6).
The 2022 iteration proposes unlocking phenotypic plasticity as a hallmark, involving disrupted differentiation through loss of transcription factors (e.g., HOXA5 in colon cancer) or fusions like PML-RARα in leukemia, allowing dedifferentiation or transdifferentiation. Nonmutational epigenetic reprogramming, an enabling characteristic, features dynamic chromatin changes (e.g., via TET demethylases under hypoxia) without DNA alterations, facilitating heterogeneity. Polymorphic microbiomes contribute molecularly through metabolites (e.g., butyrate inducing senescence) or mutagenic enzymes (e.g., colibactin from E. coli), intersecting with inflammation. Senescent cells, via the senescence-associated secretory phenotype (SASP) involving cytokines and chemokines, enhance invasion and immune modulation.
Critiques of the framework, such as those questioning its universality across all cancers, highlight that not all tumors exhibit every hallmark equally, and some capabilities may be context-dependent. For example, certain critiques argue that systemic effects or non-cell-autonomous mechanisms deserve greater emphasis. Nonetheless, the molecular focus has spurred therapeutic advances, like PARP inhibitors for genome instability or anti-PD-1 therapies for immune evasion.
| Hallmark | Key Molecular Mechanisms | Examples of Involved Genes/Proteins |
|----------|--------------------------|-----------------------------|
| Sustaining Proliferative Signaling | Activation of growth factor pathways | RAS, PI3K, EGFR |
| Evading Growth Suppressors | Inactivation of cell cycle regulators | TP53, RB1, CDKN2A |
| Resisting Cell Death | Alteration of apoptosis pathways | BCL-2, BAX, NF-κB |
| Enabling Replicative Immortality | Telomere maintenance | hTERT, ALT pathways |
| Inducing Angiogenesis | Upregulation of angiogenic factors | VEGF, HIF-1α |
| Activating Invasion and Metastasis | EMT and ECM remodeling | SNAIL, ZEB1, MMPs, TP53 |
| Reprogramming Energy Metabolism | Shift to glycolysis | GLUT1, PKM2, LDHA |
| Evading Immune Destruction | Immune checkpoint activation | PD-L1, CTLA-4, TGF-β |
| Unlocking Phenotypic Plasticity | Disrupted differentiation | HOXA5, PML-RARα, SOX9 |
| Nonmutational Epigenetic Reprogramming | Chromatin modifications | TET demethylases, H1.0 histone |
| Polymorphic Microbiomes | Metabolite and mutagen production | Colibactin (E. coli), butyrate producers |
| Senescent Cells in Microenvironment | SASP secretion | IL-6, chemokines, cytokines |
This table summarizes the hallmarks with molecular examples, illustrating their interconnectedness. Overall, the hallmarks framework continues to evolve, integrating molecular insights to advance cancer research and treatment.
### Key Citations
- [Hallmarks of Cancer: The Next Generation - Cell Press](https://www.cell.com/fulltext/S0092-8674%2811%2900127-9)
- [Hallmarks of Cancer: New Dimensions | Cancer Discovery](https://aacrjournals.org/cancerdiscovery/article/12/1/31/675608/Hallmarks-of-Cancer-New-DimensionsHallmarks-of)
- [The Hallmarks of Cancer - Cell Press](https://www.cell.com/fulltext/S0092-8674%2800%2981683-9)
- [Hallmarks of cancer: of all cancer cells, all the time? - PubMed](https://pubmed.ncbi.nlm.nih.gov/22795735/)
- [Hallmarks of cancer and hallmarks of aging - Aging-US](https://www.aging-us.com/article/204082/text)
- [Molecular principles of metastasis: a hallmark of cancer revisited](https://www.nature.com/articles/s41392-020-0134-x)
- [Embracing cancer complexity: Hallmarks of systemic disease](https://www.sciencedirect.com/science/article/pii/S0092867424001752)
Kommentare