Fact-checked by Grok 2 weeks ago

Cytostasis

Cytostasis refers to the inhibition of cell growth and division, resulting in a reversible arrest of cellular proliferation without direct cell death. In oncology, cytostatic agents are substances that slow or stop the expansion of cancer cells, often stabilizing tumor size and preventing metastasis rather than causing tumor regression. Beyond oncology, cytostasis plays essential roles in immune responses, development, and tissue homeostasis. Cytostatic therapies form a key component of modern , particularly in regimens targeting rapidly dividing malignant cells. These agents, such as antimetabolites (e.g., ) and tyrosine kinase inhibitors (e.g., ), primarily interfere with , pathways, or progression to induce cell cycle arrest in tumor cells. By halting proliferation, cytostatics can extend patient survival and reduce symptoms, though they may also affect normal proliferating tissues like and gastrointestinal , leading to side effects such as myelosuppression and . Distinguished from cytotoxic approaches that promote , cytostatic mechanisms emphasize growth suppression, which has implications for targeted therapies like tyrosine kinase inhibitors. This cytostatic effect challenges traditional endpoints focused on tumor shrinkage, necessitating alternative metrics such as to assess efficacy. Ongoing research explores combining cytostatics with immunotherapies to enhance anti-tumor responses while minimizing resistance.

Definition and Fundamentals

Definition

Cytostasis, derived from the Greek roots "cyto-" meaning cell and "stasis" meaning stoppage or standing still, refers to the reversible inhibition of , division, and proliferation without inducing . This process maintains cellular viability while halting progression through the , distinguishing it from lethal cellular responses. Key characteristics of cytostasis include the arrest of cells in specific phases of the , such as G0/G1 or /M, where is temporarily suspended but can resume upon removal of the inducing agent or stimulus. This reversibility is a hallmark feature, allowing cells to recover metabolic and replicative functions once the cytostatic condition is alleviated, thereby preventing permanent tissue damage in biological systems.

Distinction from Cytotoxicity

Cytostasis and represent distinct cellular responses to inhibitory stimuli, with cytostasis primarily halting through mechanisms such as arrest or metabolic slowdown, without inducing , while actively triggers pathways leading to , , or other forms of irreversible cell demise. This fundamental difference influences therapeutic outcomes in contexts like , where cytostatic agents aim to stabilize disease by preventing tumor expansion, in contrast to cytotoxic agents that seek tumor reduction via direct cell killing. A key implication of this distinction lies in the potential for cellular : under cytostatic conditions, viable cells can often resume normal once the inhibiting is removed, allowing for reversible growth arrest that preserves overall integrity. Cytotoxic effects, however, culminate in permanent cell loss, as the activation of pathways precludes any , often resulting in measurable damage or therapeutic gauged by tumor shrinkage. This reversibility in cytostasis supports strategies focused on long-term management, whereas cytotoxicity aligns with aggressive interventions requiring careful monitoring of side effects due to non-selective cell elimination. Distinguishing these effects experimentally relies on targeted assays that separate proliferation inhibition from viability loss. Cytostasis is commonly evaluated via proliferation-specific methods, such as the , which detects reduced metabolic activity as a proxy for slowed while confirming intact viability, or BrdU incorporation assays that quantify diminished in cycling cells without a corresponding drop in total cell count. In contrast, is assessed through death indicators like the LDH release assay, which measures efflux from cells with compromised membranes as a marker of or , or trypan blue exclusion, where dye uptake reveals non-viable cells unable to maintain membrane integrity. These complementary approaches enable precise , informing the development of agents with desired cytostatic or cytotoxic profiles.

Mechanisms of Action

Cellular Level Processes

Cytostasis primarily manifests at the cellular level through the of at key checkpoints in the , resulting in reduced during the and halted progression into or through . This prevents cells from completing division, thereby inhibiting without necessarily causing . In response to such signals, cells often transition into quiescence, a reversible G0 state where they exit the active but retain the potential to re-enter upon favorable conditions, or into senescence-like states characterized by more persistent proliferative inhibition. Observable effects of cytostasis include alterations in cell morphology, such as flattening and changes in shape, alongside accumulation of cells in the of the without the activation of damage response, distinguishing it from damage-induced arrests. In quiescent states induced by cytostatic cues, cells may exhibit compact or altered structural features, while senescence-like responses often involve enlarged, flattened morphologies with increased lysosomal activity. These changes reflect a shift toward maintenance and survival rather than growth, with no overt signs of genomic instability in non-damaging contexts. Experimental evidence from models underscores these processes, demonstrating a halt in when cells are subjected to nutrient deprivation, such as serum starvation, or contact inhibition in confluent cultures. For instance, serum withdrawal in cell lines like C3H10T1/2 leads to rapid accumulation of over 80% of cells in the G0/ within 20-48 hours, accompanied by decreased expression of markers like Ki-67, without triggering DNA damage pathways. Similarly, contact inhibition in epithelial or mesenchymal cells elevates cyclin-dependent kinase inhibitors, enforcing G1 arrest and quiescence that is reversible upon replating at lower densities, highlighting the adaptive nature of cytostatic responses to environmental constraints.

Molecular Pathways Involved

Cytostasis involves several key molecular pathways that halt without inducing , primarily through the regulation of progression and metabolic adaptation. One central pathway is the -mediated arrest, where the tumor suppressor protein responds to DNA damage or other stresses by upregulating the (CDK) inhibitor p21 (also known as CDKN1A). This upregulation inhibits CDK activity, preventing phosphorylation of the () and thereby blocking the , which enforces temporary arrest to allow . The induction by forms a feedback loop that sustains inhibition of CDKs, ensuring sustained quiescence until the stress is resolved, as demonstrated in seminal studies on -dependent G1 arrest. The pathway further reinforces cytostatic effects by acting as a at the G1/S checkpoint. In its hypophosphorylated state, binds to transcription factors, repressing genes required for S-phase entry, such as cyclins E and A. Stress signals maintain in this repressive form through upstream regulators like p16INK4a or , preventing E2F-mediated transcription and thus inhibiting without triggering . This pathway integrates with signaling, where p21-mediated CDK inhibition keeps active, creating a coordinated block that promotes reversible proliferation arrest. In response to energy stress, (AMPK) activation provides another cytostatic mechanism by sensing low ATP/AMP ratios and phosphorylating targets that suppress anabolic processes. AMPK inhibits signaling, reducing protein synthesis and while promoting catabolic pathways to restore energy balance, which collectively limits at sub-cytotoxic levels. This activation establishes a where cells enter a quiescent rather than undergoing . Transcription factors such as FOXO proteins play a pivotal role in promoting and maintaining cellular quiescence within these pathways. FOXO3, for instance, translocates to the upon stress-induced (e.g., via PI3K/Akt inhibition), where it upregulates genes like p27 and G2 to reinforce CDK inhibition and exit. FOXO factors form feedback loops with and by enhancing p21 expression and coordinating with AMPK to sustain quiescence, ensuring long-term maintenance without exhaustion. These pathways integrate with broader stress responses, notably through inhibition, which reduces global protein synthesis by preventing the of 4E-BP1 and S6K1, thereby promoting inhibition of initiation and enforcing cytostasis under nutrient or energy limitation. At cytostatic thresholds, suppression—often downstream of AMPK or —halts while preserving viability, distinguishing it from cytotoxic overload, as evidenced in profiling studies of showing correlated reductions in cell size and biosynthetic rates.

Biological Significance

Role in Immune Responses

Cytostasis plays a crucial role in immune responses by enabling effector cells to inhibit the of infected or malignant cells, thereby controlling spread or tumor expansion while minimizing collateral tissue damage. Activated macrophages and natural killer () cells are primary mediators of this process, secreting soluble factors such as tumor necrosis factor-alpha (TNF-α) and interferons that directly suppress target . For instance, monocytes stimulated with interferon-gamma (IFN-γ) exhibit enhanced cytostatic activity against cells through TNF-α-dependent mechanisms, where these cytokines arrest the without necessarily inducing . Similarly, NK cells contribute to cytostasis by releasing TNF-α and lymphotoxin, which inhibit tumor cell growth in a non-cytolytic manner, complementing their cytotoxic functions. In antiviral immunity, cytostasis limits the replication of infected cells, preventing viral dissemination. Type I interferons, produced by plasmacytoid dendritic cells and other innate immune components, exert cytostatic effects on virus-infected cells by upregulating genes that halt the cell cycle, such as those involved in double-stranded RNA sensing and signaling. This mechanism is particularly vital during early infection phases, where IFN-α restricts host cell proliferation to curb viral propagation without widespread cell death. In antitumor immunity, cytostasis curbs the metastatic potential of cancer cells by impeding their proliferative capacity. Tumor-infiltrating NK cells and macrophages collaborate to enforce this control; for example, early NK cell infiltration into tumors enhances macrophage activation, leading to sustained inhibition of cancer cell division and reduced metastasis in experimental models. Experimental in vivo studies highlight the role of macrophage-derived arginase in mediating cytostasis through L-arginine depletion. Myeloid cells, including tumor-associated macrophages, upregulate arginase-1 (ARG1) in the tumor microenvironment, which hydrolyzes L-arginine into ornithine and urea, starving proliferating T cells of this essential amino acid. This depletion induces cell cycle arrest in T lymphocytes, impairing their expansion and anti-tumor responses, thereby contributing to immune suppression and tumor progression in mouse models of cancer. In one such study, ARG1-expressing macrophages in renal cell carcinoma models suppressed T-cell proliferation via localized arginine scarcity, promoting tumor immune evasion. These findings underscore cytostasis as a finely tuned immune strategy, often linked to pathways like TNF signaling for coordinated effector responses.

Functions in Development and Homeostasis

Cytostasis plays a critical role in maintaining tissue by regulating in response to environmental cues. In epithelial tissues, contact inhibition serves as a primary mechanism to prevent overgrowth, where increased cell-cell contacts trigger signaling pathways that halt proliferation upon reaching confluence. This process is essential for preserving epithelial integrity and organ size, as demonstrated in studies of polarized epithelial cells where high density restricts TGF-β signaling to the basolateral membrane, thereby inhibiting SMAD3 activation and downstream proliferative responses. Similarly, nutrient-sensing pathways contribute to cytostasis during periods of scarcity, particularly in the liver, where inhibits activity, suppressing anabolic processes like protein synthesis and inducing a reversible arrest to prioritize catabolic and energy conservation. During , cytostasis orchestrates precise temporal control of to ensure proper patterning and termination. In embryogenesis, temporary arrest occurs in structures such as the limb bud, where senescence-like cytostasis in the apical ectodermal ridge (AER) limits FGF expression and coordinates patterning genes like GLI3 and MSX2, as evidenced by p21-mediated arrest in embryos. This programmed arrest, followed by apoptotic clearance, prevents excessive expansion and supports limb morphogenesis without permanent damage. In , cytostasis facilitates the transition from proliferative repair to resolution, particularly in fibroblasts that initially expand to deposit but subsequently enter G1-phase arrest. TGF-β signaling induces this halt by upregulating inhibitors such as p15 and p21, ensuring controlled remodeling and preventing fibrotic overgrowth. Dysregulation of these cytostatic mechanisms, such as impaired contact inhibition in epithelial layers, can lead to uncontrolled and , disrupting normal tissue balance.

Cytostatic Agents

Natural Inducers

Natural inducers of cytostasis encompass endogenous molecules and physiological conditions that halt cell proliferation without causing cell death, often through reversible mechanisms that maintain cellular viability. These inducers play crucial roles in regulating tissue homeostasis and preventing uncontrolled growth in various biological contexts. Among the key natural inducers are cytokines such as transforming growth factor-β (TGF-β), which promotes G1 phase arrest in epithelial and other cell types by upregulating cyclin-dependent kinase inhibitors like p15^INK4B, p21^CIP1, and p27^KIP1. This arrest is mediated through Smad signaling pathways that inhibit cyclin E-CDK2 activity, effectively blocking the G1/S transition. TGF-β is secreted by various cell types, including immune cells and fibroblasts, acting in a paracrine manner to impose cytostasis on neighboring proliferating cells. Withdrawal of growth factors, such as (EGF) or components, similarly triggers cytostasis by depriving cells of mitogenic signals required for progression. In the absence of EGF or , cells enter a quiescent G0 state, characterized by reduced expression and Rb protein hypophosphorylation, which prevents E2F-mediated transcription of S-phase genes. This process is commonly observed in cultured fibroblasts and epithelial cells, where serum starvation induces reversible quiescence to conserve resources during nutrient scarcity. Environmental factors like and low extracellular also serve as potent natural inducers. , often encountered in poorly vascularized tissues, induces G1 arrest via hypoxia-inducible factor-1 (HIF-1) stabilization, which upregulates p27^KIP1 and downregulates , thereby inhibiting CDK2 activity. Low environments, arising from accumulation in ischemic or tumor-like settings, similarly promote G1/S blockade by altering ion and activating stress response pathways that elevate and p21 levels. These inducers often originate from autocrine or paracrine signaling within cellular microenvironments. For instance, endothelial cells produce nitric oxide (NO) via endothelial nitric oxide synthase (eNOS), which diffuses to adjacent vascular smooth muscle cells to exert cytostatic effects by modulating cell cycle regulators such as p21 and p27, independent of cGMP pathways in some cases. This paracrine inhibition helps maintain vascular homeostasis by preventing excessive smooth muscle proliferation. Natural inducers of cytostasis are typically reversible and highly context-dependent, allowing cells to resume upon signal restoration. In stem cell niches, such as hematopoietic or compartments, quiescence induced by niche-derived factors like TGF-β or BMPs ensures long-term tissue maintenance while protecting the pool from exhaustion. This reversibility distinguishes natural cytostasis from permanent , enabling adaptive responses to fluctuating physiological demands.

Pharmacological Agents

Pharmacological agents for cytostasis encompass synthetic or semi-synthetic compounds engineered to halt cell proliferation without immediate cell death, primarily targeting rapidly dividing cells in conditions like cancer. These agents contrast with natural inducers such as cytokines by offering controlled, therapeutic dosing through pharmaceutical formulations. Major classes of cytostatic agents include antimetabolites, which interfere with DNA and RNA synthesis by mimicking essential metabolites. A prominent example is methotrexate, an analog of folic acid that inhibits dihydrofolate reductase (DHFR), disrupting folate metabolism and thereby arresting the cell cycle in S phase. Microtubule stabilizers represent another key class, binding to tubulin to prevent microtubule depolymerization and suppress mitotic spindle dynamics, leading to G2/M phase arrest. Paclitaxel exemplifies this mechanism, stabilizing microtubules and blocking mitosis in proliferating cells. Tyrosine kinase inhibitors (TKIs) form a targeted class that blocks signal transduction pathways driving uncontrolled growth; imatinib, for instance, selectively inhibits the BCR-ABL tyrosine kinase fusion protein, inducing cytostasis in chronic myeloid leukemia cells by halting proliferative signaling. The development of these agents traces back to the 1950s era of , when early compounds like emerged from research on antagonists, marking a shift toward exploiting metabolic vulnerabilities in cancer cells. Initially derived from wartime chemical agents such as nitrogen mustards, these drugs evolved from broad cytotoxic profiles to more nuanced cytostatic applications at lower doses, minimizing cell killing while emphasizing growth inhibition; targeted TKIs like , approved in 2001, further refined this specificity by addressing molecular drivers. Pharmacokinetics of cytostatic agents vary by class, influencing dosing regimens and therapeutic windows. exhibits a terminal of 3 to 10 hours at low doses, primarily eliminated via renal excretion, necessitating monitoring to avoid accumulation. , administered intravenously, has an elimination of approximately 5.8 hours for infusions, with hepatic metabolism and biliary excretion. , an oral agent, achieves steady-state concentrations with a of about 18 hours, enabling once-daily dosing through hepatic metabolism. For targeted therapies, monoclonal antibodies like , which induces cytostasis in HER2-overexpressing cells by blocking receptor signaling, demonstrate prolonged pharmacokinetics with a of approximately 28 days, facilitating less frequent administration.

Clinical Applications

Use in Cancer Treatment

Cytostatic strategies in cancer treatment aim to inhibit tumor cell proliferation rather than directly inducing cell death, thereby slowing disease progression and creating opportunities for synergy with other therapies, such as cytotoxic agents or immune-mediated clearance. By arresting cells in specific phases of the , these approaches maintain stable disease states, prevent metastatic spread, and allow the to recognize and eliminate non-proliferating tumor cells over time. For instance, hormonal therapies like exert cytostatic effects in estrogen receptor-positive by inducing G0/G1 cell cycle arrest, thereby halting tumor growth without immediate . Clinical evidence supports the efficacy of cytostatic agents, particularly in hormone receptor-positive . CDK4/6 inhibitors, such as , target the pathway to enforce G1 arrest, significantly extending when combined with endocrine therapy. In the PALOMA-2 trial, plus achieved a median of 24.8 months compared to 14.5 months with alone in postmenopausal women with hormone receptor-positive, HER2-negative advanced . The U.S. approved in 2015 based on this accelerated approval pathway, highlighting its role in first-line treatment. Similarly, tamoxifen's cytostatic mechanism has been a cornerstone of , reducing recurrence risk by inducing arrest in proliferating cells. Compared to traditional cytotoxic chemotherapies, cytostatic treatments offer advantages including greater target selectivity, which minimizes damage to normal cells and reduces severe side effects like myelosuppression. They enable continuous dosing rather than intermittent cycles, making them suitable for indolent tumors with low rates. Monitoring response focuses on and tumor markers, such as CA 15-3 or CEA in , rather than radiological tumor shrinkage, as cytostasis stabilizes rather than reduces tumor volume. This shift emphasizes long-term disease control over acute cell kill, improving patient while maintaining therapeutic pressure on the tumor.

Applications in Other Diseases

Cytostasis plays a key role in managing autoimmune diseases by inhibiting excessive immune cell proliferation. Sirolimus, an mTOR inhibitor, exerts cytostatic effects by halting T-cell proliferation, making it a cornerstone in preventing transplant rejection; clinical studies demonstrate its efficacy in renal transplantation by reducing acute rejection rates through selective immunosuppression without broadly depleting immune cells. In rheumatoid arthritis, sirolimus restores the Th17/Treg balance by upregulating regulatory T cells and inhibiting synovial fibroblast proliferation, with phase 1/2 trials showing improved disease activity scores and reduced need for additional disease-modifying antirheumatic drugs in refractory cases. Beyond , cytostatic agents address other proliferative conditions, such as (BPH), where unchecked prostate cell growth leads to urinary obstruction. , a , acts cytostatically by blocking production, thereby inhibiting epithelial and proliferation in the ; long-term trials indicate it reduces prostate volume by up to 20% and lowers the risk of acute by approximately 57% in men with enlarged prostates. Emerging applications extend to , where halting division mitigates excessive deposition and scarring. , an approved antifibrotic for , inhibits proliferation via downregulation of TGF-β1/mTOR/p70S6K signaling, with preclinical and clinical data showing reduced production and improved lung function in fibrotic models. Similar cytostatic mechanisms are under investigation for cardiac and intestinal , where suppresses differentiation and in a concentration-dependent manner. Clinical trials in the 2020s have highlighted cytostatic biologics for , a condition driven by overgrowth. Bimekizumab, a dual IL-17A/IL-17F inhibitor approved by the FDA in 2023, controls hyperproliferation by neutralizing proinflammatory cytokines that drive epidermal turnover; phase 3 trials (BE READY and BE VIVID) reported that 85-91% of patients achieved at least 90% improvement in scores at week 16, with sustained efficacy through 52 weeks and high rates of complete skin clearance. This approval underscores the shift toward targeted cytostatic therapies that modulate immune-driven proliferation without broad , offering superior outcomes in moderate-to-severe cases compared to earlier monotherapies.

Research Developments

Key Historical Milestones

The concept of cytostasis, referring to the reversible inhibition of cell proliferation without cell death, began to take shape in the mid-20th century through studies on the effects of ionizing radiation on cells. In the 1940s and 1950s, researchers investigating radiation therapy for cancer observed that sublethal doses could halt cell division in various tissues, laying the groundwork for understanding non-lethal growth arrest mechanisms, as seen in early experiments with X-rays on mammalian cells. A pivotal demonstration came in 1961, when sublethal doses of X-ray radiation were shown to induce a transient G2 phase premitotic block in HeLa cells, highlighting radiation's capacity to trigger cell cycle checkpoints without causing immediate lethality. The 1970s and early 1980s marked the identification of soluble factors capable of inducing cytostasis, with (TGF-β) emerging as a key player. Initially isolated in 1978 as sarcoma growth factor from tumor cells, TGF-β was later characterized for its dual role in proliferation. By 1985, studies confirmed TGF-β as a potent cytostatic agent, potently inhibiting the growth of normal and transformed epithelial cells by arresting them in the of the through induction of inhibitors. In the , the development of targeted pharmacological agents began to exploit cytostatic mechanisms, shifting from broad cytotoxics to more selective inhibitors. Early examples included interferon-alpha, approved by the FDA in 1986 for , which exerted cytostatic effects by modulating pathways to suppress in hematopoietic malignancies. This era also saw the groundwork for monoclonal antibody-based therapies, with initial preclinical successes in targeting receptors to induce arrest in solid tumors. The 2010s witnessed the integration of cytostatic agents with , enhancing antitumor responses by combining proliferation blockade with immune activation. Checkpoint inhibitors like , approved in 2014 for , were increasingly combined with cytostatics such as CDK4/6 inhibitors; for instance, received FDA approval in 2015 for HR-positive , and subsequent trials explored its synergy with PD-1/PD-L1 blockers to prevent tumor escape via slowed growth. These combinations demonstrated improved in clinical settings, capitalizing on cytostasis to prolong immune-mediated tumor control. As of 2025, advancements in cytostatic therapies include expanded approvals for next-generation CDK4/6 inhibitors in earlier disease stages. In September 2024, the FDA approved plus endocrine therapy for adjuvant treatment of high-risk HR-positive, HER2-negative early , based on the NATALEE trial showing a 25.1% reduction in invasive disease-free survival events, underscoring cytostasis's role in preventing recurrence. Recent research highlights the integration of cytostatic agents with therapies to enhance the cytostatic hold on tumor cells, particularly in solid tumors. By preconditioning with cytostatic chemotherapies such as and , immunosuppressive cells like regulatory T cells and myeloid-derived suppressor cells are depleted, facilitating greater CAR-T cell infiltration and persistence. Clinical trials, including NCT03874897, have demonstrated response rates of up to 75% in pretreated patients, underscoring this combinatorial approach as a promising trend for overcoming barriers. Post-2020 investigations have increasingly elucidated the role of cytostasis in the (SASP), a secretory program triggered by persistent that exerts paracrine effects on surrounding tissues. In cancer, cytostasis-induced SASP can initially suppress tumorigenesis by recruiting immune effectors via chemokines like but often promotes progression, , and therapy resistance through proinflammatory cytokines such as IL-6 and TGF-β. In aging contexts, cytostasis sustains SASP-mediated chronic inflammation and tissue dysfunction, exacerbating pathologies like and , as evidenced by studies showing reduced SASP upon senescent cell clearance in dermal fibroblasts. These findings have spurred therapeutic modulation of SASP, including senomorphics like rapamycin to attenuate harmful secretions without eliminating cytostatic cells. A major challenge in cytostasis-based therapies remains the development of resistance through pathway reactivation, where inhibited signaling cascades are bypassed via compensatory mechanisms. For instance, in non-small cell lung cancer treated with inhibitors, mutations like BRAF V600E reactivate the MAPK pathway, restoring proliferative signals despite initial cytostasis. Similarly, KRAS G12C inhibitors encounter resistance via -mediated ERK1/2 reactivation, highlighting the need for multi-pathway targeting. Toxicity to normal cells poses another hurdle, particularly through long-term (ROS) induction by cytostatics. A 2025 review details how agents like elevate in cardiomyocytes, leading to via oxidative damage, with selective interventions mitigating effects in preclinical models. This off-target ROS accumulation risks systemic issues like , complicating chronic dosing regimens. Looking ahead, (AI)-driven is emerging to develop selective cytostatic inducers that spare normal cells. AI-driven has identified novel inhibitors targeting oncogenic drivers like STK33, achieving selective cytostasis and tumor reduction in models. Such tools prioritize specificity by integrating multi-omics data for pathway prediction. As of 2025, clinical trials are exploring cytostasis modulation in aging-related diseases, often through -targeted interventions that address cytostatic states. For example, the combination of and has shown safety and preliminary efficacy in reducing SASP markers in (NCT02874989) and diabetic kidney disease (NCT02848131), with ongoing studies in (NCT04063124) evaluating impacts on cognitive senescence. These efforts aim to harness cytostasis for therapeutic benefit while mitigating pathological persistence.

References

  1. [1]
    Definition of cytostatic agent - NCI Dictionary of Cancer Terms
    A substance that slows or stops the growth of cells, including cancer cells, without killing them. These agents may cause tumors to stop growing and spreading.Missing: biology | Show results with:biology
  2. [2]
    Cytostasis - an overview | ScienceDirect Topics
    Cytostasis is defined as a state of halted cell division, which can be assessed through the nuclear division index in the context of cellular responses to ...
  3. [3]
    The role of cytostatic in oxidative stress reactions - PMC - NIH
    Oct 13, 2025 · Cytostatic drugs are widely applied in cancer therapy. Among the most commonly used agents are anthracyclines, such as doxorubicin, and platinum ...
  4. [4]
    Cytostatic Drugs as a Weapon against Cancer - TherapySelect
    Cytostatic drugs slow or stop cell growth by inhibiting cell division, primarily used in cancer treatment to fight fast-spreading tumor cells.
  5. [5]
    How do cytostatic agents work? - GPOH
    Feb 17, 2022 · The anticancer effect of a cytostatic drug is provided by its mechanism of action. Different cytostatics show different properties and modes of action.<|control11|><|separator|>
  6. [6]
    Is Cell Death a Critical End Point for Anticancer Therapies or Is ...
    Dec 19, 2007 · At the cellular level, cytostasis may be defined as the inhibition of cell growth and/or proliferation, and this initial event can be followed ...<|control11|><|separator|>
  7. [7]
    Sustained oncogenic signaling in the cytostatic state enables ... - NIH
    Many advanced therapeutics possess cytostatic properties that suppress cancer cell growth without directly inducing death. Treatment-induced cytostatic ...
  8. [8]
    CYTOSTASIS Definition & Meaning - Dictionary.com
    Cytostasis definition: arrest of cellular growth and division.. See examples of CYTOSTASIS used in a sentence.Missing: biology | Show results with:biology
  9. [9]
    Cytostasis - an overview | ScienceDirect Topics
    Cytostasis is defined as the inhibition of tumor cell growth, mediated by immune cells such as activated macrophages and characterized by reduced target ...
  10. [10]
  11. [11]
  12. [12]
    Drug development in oncology: classical cytotoxics and molecularly ...
    This paper will review the strategies used in exploratory trials of conventional anticancer drugs and introduce emerging paradigms for the development of novel ...
  13. [13]
    Modeling Cytostatic and Cytotoxic Responses to New Treatment ...
    A comparative preclinical study of several treatment regimens for ovarian cancer with paclitaxel, cisplatin, and bevacizumab alone or in combination.<|control11|><|separator|>
  14. [14]
    Cell-Based Methods for Determination of Efficacy for Candidate ...
    Sep 22, 2018 · One potential benefit of the LDH cytotoxicity assay is the potential to distinguish between cell death and growth inhibition with a modified LDH ...
  15. [15]
  16. [16]
    Cytotoxicity Assays: In Vitro Methods to Measure Dead Cells - NCBI
    May 1, 2019 · Cytotoxicity assays measure dead cells by detecting loss of membrane integrity, using markers that leak or dyes that penetrate damaged ...
  17. [17]
    Understanding Cytotoxicity and Cytostaticity in a High-Throughput ...
    Although well studied from a biological viewpoint, the links between biological (cytotoxic and cytostatic) space and chemical space are poorly understood.Missing: earliest | Show results with:earliest
  18. [18]
    Cellular Mechanisms and Regulation of Quiescence - PMC
    Quiescence is a state of reversible proliferative arrest in which cells are not actively dividing, and yet retain the capacity to reenter the cell cycle.
  19. [19]
    Mechanisms of Cellular Senescence: Cell Cycle Arrest ... - Frontiers
    Cell cycle arrest can be an alarm response instigated by aberrant proliferation or deleterious stress stimuli to prevent the propagation of dysfunctional cells.Missing: experimental | Show results with:experimental
  20. [20]
    Cellular quiescence induced by contact inhibition or serum ... - NIH
    These results indicate that contact inhibition and serum withdrawal lead to cellular quiescence through distinct genetic and molecular mechanisms.Missing: morphology size
  21. [21]
    p53-mediated G1 arrest requires the induction of both p21 and Killin ...
    Dec 8, 2021 · p53 induces G1 arrest primarily through activation of its best-characterized downstream gene, p21, which encodes a cyclin-dependent kinase (CDK) ...
  22. [22]
    Cell cycle regulation: p53-p21-RB signaling - Nature
    Mar 31, 2022 · Classical RB-E2F target genes often control the cell cycle by contributing to DNA replication and the transition from the G1 to S phase.Missing: cytostasis | Show results with:cytostasis
  23. [23]
    Inhibition of DNA synthesis by RB: effects on G1/S transition ... - NIH
    The ability of RB to inhibit the G1/S transition is well established by multiple lines of evidence. Physiological growth inhibitors such as TGF-β inhibit RB ...Missing: cytostasis | Show results with:cytostasis
  24. [24]
    AMPK - a nutrient and energy sensor that maintains energy ...
    Consistent with a role in maintaining energy homeostasis, when AMPK is activated by energetic stress it switches on catabolic pathways that generate ATP, while ...
  25. [25]
    Discrete mechanisms of mTOR and cell cycle regulation by AMPK ...
    Both AMPK agonists inhibited proliferation, but through unique AMPK-independent mechanisms and both reduced tumor growth in vivo independent of AMPK.Results · Active Ampk Is Abundantly... · Ampk-Dependent Mtor...
  26. [26]
    FOXO3 Promotes Quiescence in Adult Muscle Stem Cells during the ...
    Mar 20, 2014 · We demonstrate that FOXO3, perhaps by activating Notch signaling, promotes the quiescent state during SC self-renewal in adult muscle regeneration.
  27. [27]
    Review FoxO Transcription Factors and Stem Cell Homeostasis
    Aug 16, 2007 · Several recent studies indicate that FoxO-dependent signaling is required for long-term regenerative potential of the hematopoietic stem cell (HSC) compartment.
  28. [28]
    Protein Profiling Identifies mTOR Pathway Modulation and Cytostatic ...
    Consistent with mTOR inhibition, there was also a reduction in protein synthesis that correlated with cell size reduction and growth inhibition with AZD1208 ...
  29. [29]
    Role of interferon gamma and tumour necrosis factor alpha in ...
    Role of interferon gamma and tumour necrosis factor alpha in monocyte-mediated cytostasis and cytotoxicity against a human histiocytic lymphoma cell line.
  30. [30]
    Tumor Necrosis Factor and Lymphotoxin Secretion by ... - PubMed
    Sep 15, 1988 · NK cells mediate their cytotoxicity against tumor cells through abroad array of cytotoxic and cytostatic proteins.
  31. [31]
    Role of natural interferon-alpha producing cells ... - PubMed
    The type I interferons (IFNs) have antiviral, cytostatic and prominent immunomodulatory effects, which all are of great importance during viral infections.
  32. [32]
    Early-appearing tumor-infiltrating natural killer cells play ... - PubMed
    Both natural killer (NK) cells and macrophages are thought to be the main effectors responsible for early antitumor defense. In this study, we investigated ...
  33. [33]
    identification of the soluble factors inducing cytotoxicity and the L ...
    Simultaneous treatment with polymyxin B, TNF alpha antibody, and IFN gamma antibody reduced EMT-6 cell nitrite production by 81%, and cytostasis by 74%. By ...
  34. [34]
  35. [35]
  36. [36]
  37. [37]
    TGF-β signaling in health, disease and therapeutics - Nature
    Mar 22, 2024 · For most cells, TGF-β inhibits their proliferation, or in other words, triggers their cytostasis by inducing cell cycle arrest in the gap 1 (G1) ...
  38. [38]
    Actomyosin contractility provokes contact inhibition in E-cadherin ...
    Apr 13, 2017 · Confluence-dependent inhibition of epithelial cell proliferation, termed contact inhibition, is crucial for epithelial homeostasis and organ ...
  39. [39]
    Transforming growth factor beta-induced cell cycle arrest of human ...
    Oct 19, 2004 · Up-regulation of p57 mRNA and protein occurs before TGFbeta-induced G1 cell cycle arrest, requires transcription, and is mediated via a highly conserved region ...
  40. [40]
    The TGF-beta paradox in human cancer: an update - PubMed - NIH
    TGF-beta plays an essential role in maintaining tissue homeostasis through its ability to induce cell cycle arrest, differentiation and apoptosis, ...
  41. [41]
    Starvation activates ECM-remodeling gene transcription and ...
    Jun 24, 2025 · Nutrient and growth factor depletion induces cellular quiescence, which is traditionally considered a transcriptionally inactive state. Secchia.
  42. [42]
    Hypoxia Inhibits G1/S Transition through Regulation of p27 Expression
    Hypoxia Induces a G1 Cell Cycle Arrest in Immortalized Fibroblasts and Is Associated with Hypoxia-mediated Changes in G1 Regulators. Since contact inhibition ...
  43. [43]
    Effect of Low pH Treatment on Cell Cycle and Cell Growth
    Aug 6, 2025 · In conclusion, low pH treatment affects cell growth potentially through G1 cell cycle arrest. Future work is to confirm cell cycle arrest at G1 ...Missing: cytostasis | Show results with:cytostasis
  44. [44]
    Nitric oxide modulates expression of cell cycle regulatory proteins
    NO inhibits VSMC proliferation by specifically changing the expression and activity of cell cycle regulatory proteins, which may occur independent of cGMP.
  45. [45]
    Stem cell quiescence: the challenging path to activation | Development
    Feb 8, 2021 · Quiescent adult stem cells can reversibly transition into an active state in which they enter the cell cycle and generate new differentiated ...Missing: inducers cytostasis
  46. [46]
    Mechanisms, Hallmarks, and Implications of Stem Cell Quiescence
    Jun 11, 2019 · Quiescence is a reversible G0 state, because cells retain the ability to re-enter G1 of the cell cycle after passing the restriction point (R- ...Missing: natural cytostasis
  47. [47]
    Therapeutic drug monitoring for cytotoxic anticancer drugs
    Dec 8, 2022 · Cytotoxic drugs are the oldest class of anticancer drugs. They are ... Potential drug interactions with busulfan. Drug class and agents ...
  48. [48]
    Methotrexate - StatPearls - NCBI Bookshelf - NIH
    Dec 11, 2024 · This activity reviews the indications, mechanism of action, and contraindications for methotrexate as a treatment for neoplastic and autoimmune ...
  49. [49]
    Exploring tubulin-paclitaxel binding modes through extensive ...
    Mar 11, 2025 · Specifically, paclitaxel binding to microtubules inhibits their depolymerization, thereby interfering with normal dynamics of these polymers, ...
  50. [50]
    Long-Term Outcomes of Imatinib Treatment for Chronic Myeloid ...
    Mar 9, 2017 · Imatinib, a selective BCR-ABL1 kinase inhibitor, improved the prognosis for patients with chronic myeloid leukemia (CML).
  51. [51]
    Discovery – Methotrexate: Chemotherapy Treatment for Cancer - NCI
    Apr 30, 2014 · In the 1950s and 1960s, the use of methotrexate was just beginning to be explored. It would soon prove useful in treating breast cancer ...
  52. [52]
    A History of Cancer Chemotherapy - AACR Journals
    Oct 30, 2008 · The use of chemotherapy to treat cancer began at the start of the 20th century with attempts to narrow the universe of chemicals that might affect the disease.
  53. [53]
    Paclitaxel Monograph for Professionals - Drugs.com
    Jul 17, 2025 · Half-life. Conventional paclitaxel: Average elimination half-life: 5.8 hours for 6- to 24-hour infusions, 2.33 hours for 3-hour infusions.
  54. [54]
    Clinical pharmacokinetics of imatinib - PubMed
    The terminal elimination half-life is approximately 18 hours. Imatinib plasma concentrations predictably increase by 2- to 3-fold when reaching steady state ...
  55. [55]
    Trastuzumab - StatPearls - NCBI Bookshelf - NIH
    Jun 22, 2024 · Trastuzumab is typically given in combination with one or more cytotoxic agents, most commonly anthracyclines (eg, doxorubicin) and taxanes (eg, ...
  56. [56]
    Effects of the antioestrogen tamoxifen on the cell cycle kinetics of the ...
    Jun 1, 1984 · Cells were not only arrested in the G1 phase but also in the G2 phase of the cell cycle. A few colonies of MCF-7 cells were resistant to 10 days ...
  57. [57]
    Palbociclib in Hormone-Receptor–Positive Advanced Breast Cancer
    Jun 1, 2015 · We assessed the efficacy of palbociclib (an inhibitor of CDK4 and CDK6) and fulvestrant in advanced breast cancer. Methods. This phase 3 study ...
  58. [58]
    Palbociclib (IBRANCE Capsules) - FDA
    Feb 22, 2016 · In February 2015, FDA granted accelerated approval for palbociclib in combination with letrozole for the treatment of HR-positive, HER2 ...
  59. [59]
    Low-Dose Sirolimus Immunoregulation Therapy in Patients ... - PMC
    Nov 3, 2019 · Sirolimus, an mTOR inhibitor, exhibits immunosuppressive effects via inhibition of B cell and T cell proliferation. Therefore, it was initially ...
  60. [60]
    Mechanistic target of rapamycin (mTOR): a potential new therapeutic ...
    Oct 2, 2023 · The mechanistic target of rapamycin (mTOR) signaling pathway associated with RA connects immune and metabolic signals, which regulates immune cell ...
  61. [61]
    Sirolimus Treatment in Patients with Refractory Rheumatoid Arthritis
    Sirolimus treatment selectively up-regulated Tregs and partly decreased the usage of DMARDs without over-treatment and evaluated side effect.
  62. [62]
    Finasteride targets prostate vascularity by inducing apoptosis and ...
    This evidence supports that finasteride has apoptotic and anti-angiogenic effects against benign and malignant prostate.
  63. [63]
    Pirfenidone Inhibits Cell Proliferation and Collagen I Production of ...
    Mar 22, 2020 · Pirfenidone inhibits the proliferation of intestinal fibroblasts and suppresses collagen I production through the TGF-β1/mTOR/p70S6K signaling pathway.
  64. [64]
    Pirfenidone affects human cardiac fibroblast proliferation and cell ...
    Feb 17, 2023 · Our data demonstrate that PFD inhibits human CF proliferation in a concentration-dependent manner with an IC 50 of 0.43 mg/ml.<|control11|><|separator|>
  65. [65]
    BIMZELX® Approved by the U.S. FDA for the Treatment of Adults ...
    Oct 18, 2023 · The US Food and Drug Administration (FDA) has approved BIMZELX (bimekizumab-bkzx) for the treatment of moderate-to-severe plaque psoriasis in adults.Missing: keratinocyte | Show results with:keratinocyte
  66. [66]
    Full article: Review of bimekizumab in the treatment of psoriasis
    Psoriasis is a chronic inflammatory disease which leads to uncontrolled keratinocyte differentiation and proliferation. Injury to keratinocytes causes ...
  67. [67]
    Historical view of the effects of radiation on cancer cells - PMC - NIH
    Apr 30, 2025 · This historical review traces the evolution of radiobiological theories and their impacts on current therapeutic strategies.
  68. [68]
    The Discovery and Early Days of TGF-β: A Historical Perspective
    This review summarizes the discovery of TGF-β, the early developments in its molecular and biological characterization with its many biological activities.Missing: cytostatic | Show results with:cytostatic
  69. [69]
    Evolution of Cancer Pharmacological Treatments at the Turn of the ...
    The first revolutionary pharmacological approach was represented by the use of chemotherapeutic antitumor drugs, which cytotoxic against various tumors; however ...
  70. [70]
    Rise of Antibody-Drug Conjugates: The Present and Future
    May 25, 2023 · ... first ADC trials underway in the 1980s. The first approved ADC was gemtuzumab ozogamicin in 2000, where a CD33 antibody is conjugated to ...Missing: cytostatic | Show results with:cytostatic
  71. [71]
    FDA approves Novartis Kisqali® to reduce risk of recurrence in ...
    Sep 17, 2024 · FDA approves Novartis Kisqali® to reduce risk of recurrence in people with HR+/HER2- early breast cancer. Sep 17, 2024.
  72. [72]
    Combining chemotherapy with CAR-T cell therapy in treating solid ...
    Mar 6, 2023 · This review will summarise the main effects of chemotherapy when combined with CAR-T cells, including the promises and challenges of combination therapy for ...
  73. [73]
  74. [74]
  75. [75]
  76. [76]
  77. [77]
  78. [78]
  79. [79]
  80. [80]
  81. [81]
  82. [82]
  83. [83]