scispace - formally typeset
Search or ask a question

What is the mechanism of the tumor-to-tumor carcinogenesis theory in heterogenic tumors? 


Best insight from top research papers

The tumor-to-tumor carcinogenesis theory in heterogenic tumors involves various mechanisms proposed in different studies. One theory suggests that cancer heterogeneity arises from epigenetic sources, leading to the emergence of multiple phenotypic states and kinetic paths from normal to cancer states, reflecting wider protein concentration fluctuations and weaker stability . Another hypothesis introduces the neosis paradigm, where cells escape senescence through neosis, producing a population of Tumor Repopulating Raju Cells (TRRCs) with transient stem cell properties, contributing to tumor heterogeneity . Additionally, the feedback loop model proposes that mutated genes act in a cell-heteronomous fashion, interacting reciprocally with stromal cells to form the cancer phenotype, contrary to the somatic mutation theory . Furthermore, the dysdedifferentiation process in metazoans, influenced by epigenetic mechanisms, can lead to a neoplastic phenotype, emphasizing the role of epigenetic changes in carcinogenesis .

Answers from top 5 papers

More filters
Papers (5)Insight
The neosis paradigm of carcinogenesis (NPC) proposes that tumor cells escape senescence via neosis, producing Tumor Repopulating Raju Cells (TRRCs) with transient stem cell properties, contributing to tumor heterogeneity.
Not addressed in the paper.
Not addressed in the paper.
The "feedback loop model" suggests that tumor-stromal cell interactions, altered intercellular communication, and anomalous signal processing contribute to tumor development in a cell-heteronomous manner.
Open accessJournal ArticleDOI
Cong Chen, Jin Wang, Jin Wang 
08 Feb 2016-Scientific Reports
30 Citations
The tumor-to-tumor carcinogenesis theory in heterogenic tumors is driven by epigenetic influences on gene regulation dynamics, leading to diverse phenotypic states and increased instability within the cancer cells.

Related Questions

What is tumor to tumor metastasis?4 answersTumor-to-tumor metastasis is a rare phenomenon where primary tumor cells spread hematogenously to another tumor. This unique occurrence involves one distinct malignancy within the substance of another independent tumor, making it challenging to detect and classify due to conflicting terminology in the literature. Examples include renal cell carcinoma metastasizing into a pancreatic neuroendocrine tumor, lung cancer metastasizing to a thymoma, and prostatic adenocarcinoma metastasizing to myxoid liposarcoma. Understanding tumor-to-tumor metastasis is crucial for accurate diagnosis and treatment decisions, as it can impact the progression of both primary and secondary tumors. This rare occurrence underscores the complexity of cancer metastasis and the need for careful consideration in managing such cases.
What are the proofs that there is itnra-tumor heterogneiety ?5 answersIn the field of tumor biology, there is evidence of intratumor heterogeneity, which refers to the presence of diverse cell populations within a single tumor. This heterogeneity can be observed at various levels, including genetic, epigenetic, and phenotypic differences among tumor cells. Several studies have provided proofs of intratumor heterogeneity, highlighting its importance in tumor progression, treatment response, and the development of drug resistance. For example, Fletcher et al. discuss statistical and epidemiological tests used in research papers, which can help identify and analyze heterogeneity in tumor samples. Additionally, Patsopoulos et al. emphasize the need for consistent terminology and standardized approaches to quantify and interpret heterogeneity in meta-analyses. These studies, along with others, contribute to our understanding of intratumor heterogeneity and its implications in cancer research and treatment.
What are the mechanisms of cancer?4 answersCancer is caused by genetic and epigenetic changes that disrupt the normal regulation of cell division and cell death. The development of cancer involves the activation of oncogenes and the inactivation of tumor suppressor genes. Carcinogenesis is a complex process that begins with cellular DNA mutations, which are often irreversible, and is followed by a long period of promotion that can be reversible in the early stages. The key events in carcinogenesis are primarily epigenetic and involve the regulation of gene expression through various mechanisms. Transcription factors play a crucial role in gene expression and cell identity, and their dysregulation contributes to cancer progression. Carcinogenesis can be induced by both DNA-reactive (genotoxic) carcinogens, which directly damage DNA, and epigenetic (nongenotoxic) carcinogens, which affect cellular processes involved in growth control and cell death. Understanding these mechanisms is important for developing effective cancer therapies and conducting human risk assessments.
How does the two hit hypothesis explain the development of cancer?5 answersThe two-hit hypothesis explains the development of cancer by proposing that both genetic and epigenetic mechanisms can lead to the inactivation of tumor suppressor genes. This hypothesis suggests that the first hit is a germline mutation or polymorphism that increases the risk of developing cancer, while the second hit is a somatic alteration that further promotes tumorigenesis. The first hit reduces the number of somatic alterations required for tumor development, leading to earlier diagnosis and fewer driver gene mutations. The second hit can be caused by factors such as DNA methylation in the promoter region of a gene, leading to loss of protein expression and mismatch repair deficiency. Additionally, early life stress has been proposed as a first wave hit that can impact the immune, endocrine, and cognitive systems, potentially contributing to the development and progression of breast cancer as a second wave hit.
How does clonal heterogeneity have a role in the progression of cancer?5 answersClonal heterogeneity plays a role in the progression of cancer by contributing to tumor evolution, therapeutic resistance, and immune evasion. Tumors with high intratumor heterogeneity (ITH) are associated with decreased immunosurveillance and reduced response to immune checkpoint inhibition. In certain tissues, only a rare subset of cells has the capacity to initiate cancer, while others stall upon acquiring oncogenic mutations. Genetic heterogeneity is observed between primary and metastatic lesions, as well as among multiple metastases, in colon cancer with liver metastases (CCLM). Colorectal cancer (CRC) exhibits marked heterogeneity, with an initial explosion of genetic diversity followed by a period of neutral dynamics. Thyroid cancer (TC) also demonstrates clonal selection and metastatic origin, with specific subclones colonizing distant organs. These findings highlight the importance of clonal heterogeneity in cancer progression, treatment response, and the development of metastasis.
What are the molecular mechanisms that drive tumor heterogeneity in cervical cancer?4 answersTumor heterogeneity in cervical cancer is driven by multiple molecular mechanisms. Infection by high-risk human papillomavirus (HPV) and the integration of the HPV genome into the host chromosome of cervical epithelial cells are key early events in neoplastic progression. The viral oncoproteins E6 and E7 play a crucial role in disrupting the DNA repair mechanisms and apoptosis, leading to rapid cell proliferation. Multiple genes involved in DNA repair, cell proliferation, growth factor activity, angiogenesis, and mitogenesis become highly expressed in cervical intraepithelial neoplasia (CIN) and cancer, contributing to genomic instability and progression towards invasive carcinoma. These molecular events result in the development of intratumor heterogeneity, which is characterized by the presence of divergent gene and protein expression programs in high-grade meningiomas. The expansion of sub-clonal copy number variants and alterations in immune infiltration, MAPK signaling, PI3K-AKT signaling, and cell proliferation drive meningioma recurrence. Epigenetic modifications, including reversible histone modifications and DNA methylation patterns, also contribute to drug resistance and the selection of drug-resistant tumor cells in cervical cancer.

See what other people are reading

What are the most common misconceptions among patients regarding the current treatment options for cystic fibrosis (CF)?
5 answers
Patients with cystic fibrosis (CF) may have misconceptions about the current treatment options. One common misconception could be the belief that therapies only target symptoms rather than the underlying genetic defect in CFTR, as highlighted in. Additionally, patients might not be aware of the diverse range of treatment modalities available, including small molecule CF modulators, gene therapies like CRISPR/Cas9, and potential stem cell-based treatments, as discussed in. Furthermore, there could be a misconception regarding the limited efficacy of traditional treatments like pancreatic enzyme replacement and respiratory physiotherapy, without considering the advancements in mutation-specific therapies that aim to restore CFTR function, as mentioned in. Educating patients about these evolving treatment options can help dispel misconceptions and empower them to make informed decisions about their care.
What are the recent updates in terms of exoscheletons?
5 answers
Recent updates in exoskeletons have focused on their potential therapeutic applications in various fields. Stem cell-derived exosomes have emerged as effective therapeutic vehicles due to their ability to influence cell functions like proliferation, migration, and angiogenesis. Exosomes have been explored for their role in cancer immunotherapy, where they activate the immune system and transfer molecular components to regulate immune responses. Additionally, exosomes have been studied for their involvement in inducing drug resistance in tumors and promoting metastasis. These findings highlight the diverse applications of exosomes in regenerative medicine, cancer therapy, and immune modulation, showcasing their promising role in future therapeutic interventions.
Is nanotechnology the future of treatment of brain cancer?
5 answers
Nanotechnology holds immense promise in revolutionizing the treatment of brain cancer. By leveraging nanoscale manipulation, nanoparticles can be engineered to bypass the blood-brain barrier, delivering therapeutic agents precisely to brain tumors. This targeted approach minimizes damage to healthy tissues and enhances treatment efficacy, addressing the limitations of conventional therapies like chemotherapy and radiotherapy. Nanotechnology also offers the potential for precise drug delivery, reducing toxic side effects and improving drug half-life and selectivity. Despite challenges like nanoparticle toxicity and biocompatibility, ongoing research aims to optimize nanotechnology's effectiveness in treating brain tumors. Therefore, the advancements in nanotechnology-based strategies signify a promising future for brain cancer therapy, emphasizing the potential of nanomedicine in enhancing treatment outcomes.
Cancer of epithelial lineage
5 answers
Cancer of epithelial lineage refers to tumors originating from epithelial cells, which are common in various cancers like breast, prostate, lung, and bladder cancers. These cancers often exhibit lineage plasticity, where cells switch between different developmental pathways, impacting tumor progression, metastasis, and therapy resistance. For instance, breast cancer subtypes retain metabolic features of their putative cell of origin, highlighting the importance of understanding normal mammary lineage metabolic identities in cancer therapy. In prostate cancer, lineage plasticity can be quantified through measurable biomarker signatures, aiding in assessing the effects of therapies on this property. Additionally, bladder cancer cell lines like JAM-ICR exhibit characteristics of both mesenchymal and epithelial lineages, providing insights into the molecular mechanisms of bladder cancer pathogenesis and drug evaluation.
Is there tissue specification in the context of immune cells, inflammation and wound healing? are there tissue specific haemocytes?
5 answers
Tissue specificity plays a significant role in immune cell distribution, inflammation, and wound healing processes. Studies have shown that immune cells adapt to local microenvironments, acquiring distinct features and functional specializations across different tissues. Additionally, chemokine receptors are implicated in the tissue-specific homing of stem cells, suggesting a role in tissue-specific homing prior to differentiation. In the context of wound healing, leukocytes, particularly macrophages, are crucial for stimulating the proliferative phase of healing through the production of growth factors that support cell proliferation and protein synthesis. Furthermore, the complement system, with tissue-specific regulation of complement gene expression, is essential in inflammation and local host defenses, highlighting the importance of tissue-specific responses in immune processes. These findings collectively indicate the existence of tissue-specific adaptations in immune cells and haemocytes.
What is the link between senescence, fibrosis and cancer ?
5 answers
Senescence plays a crucial role in connecting fibrosis and cancer. Senescent cells develop a senescence-associated secretory phenotype (SASP) that includes the secretion of cytokines, growth factors, and exosomes, promoting fibrosis. This SASP can induce genetic instability and epithelial-mesenchymal transition, facilitating cancer progression. Additionally, senescent fibroblasts acquire a cancer-associated fibroblast (CAF) phenotype, promoting tumor growth and metastasis through the secretion of specific factors and increased collagen activity. The interplay between senescence, fibrosis, and cancer involves shared mechanisms such as TGF-β signaling, oxidative stress, and cellular senescence, contributing to the development and progression of both fibrotic diseases and cancers. Understanding these connections is crucial for developing innovative treatment strategies targeting common pathways in fibrosis and cancer.
How does cell therapy work in terms of introducing new cells into tissues?
5 answers
Cell therapy involves introducing new cells into tissues to treat diseases. This process can include various methods such as transplanting stem cells, mature functional cells, modified human cells, xenotransplantation, or trans-differentiated cells. The cells used in therapy can be sourced from the patient, a donor, or another species. Stem cells, capable of self-renewal and differentiation, are commonly used for tissue repair and repopulation. To ensure the success of cell therapy, factors like donor qualification, sterile environments, and product purity must be considered. Novel techniques like in vivo cell conversion have been developed to utilize endogenous cells for tissue repair, reducing the need for external cell sources and potential immunorejection risks. Overall, cell therapy aims to replace, repair, or enhance damaged tissues or organs using carefully selected and manipulated cells.
What are the molecular mechanisms involved in the development of zebrafish eyes?
6 answers
The development of zebrafish eyes involves a complex interplay of molecular mechanisms, beginning with the specification of primordial germ cells and their migration, which is crucial for the formation of gametes necessary for the next generation of zebrafish. This process is facilitated by germinal granules and components like Dead end, which are essential for germ cell development, including motility and fate maintenance. The DNase domain-containing protein TATDN1 plays a significant role in eye cell development by ensuring proper chromosome segregation, with its deficiency leading to disordered eye cell layers and smaller eyes due to abnormal cell cycle progression and polyploidy. The oncoprotein SET/I2PP2A, through its expression in the eye, otic vesicle, brain, and lateral line system, influences cell proliferation and apoptosis, impacting the sensory system development, including the retina. The vertebrate lens development in zebrafish, characterized by rapid differentiation of epithelial cells and denucleated fiber cells, is regulated by gene expression changes, highlighting the importance of α-crystallins, β-crystallins, and γ-crystallins in lens development. Neuronal targeting and lamination within the zebrafish visual system, including the inner plexiform layer of the retina and the synaptic region of the optic tectum, are governed by molecules identified through genetic screens, emphasizing the role of cell-cell interactions and specific RNA helicases in neurite targeting and sublamination. The development and maintenance of photoreceptors are critically dependent on intracellular trafficking, with proteins like Rab8 and the exocyst complex playing central roles in ciliogenesis and the trafficking of ciliary proteins to the photoreceptor cell. Lastly, the retinal homeobox (Rx) transcription factors, particularly Rx3, regulate a network of genes during early eye morphogenesis, influencing optic vesicle morphogenesis and repressing brain development through modulated expression of genes involved in Wnt signaling and retinoid-signaling pathways. Collectively, these molecular mechanisms underscore the intricate regulatory networks governing zebrafish eye development, from germ cell specification to the differentiation of retinal neurons and lens cells, and the establishment of functional photoreceptors.
How to reduce nitric oxide levels in the colon?
5 answers
To reduce nitric oxide (NO) levels in the colon, various approaches can be considered based on the data from the provided contexts. Natural agents like polyphenols and flavonoids, as well as synthetic molecules such as dexamethasone, pioglitazone, and tropisetron, have shown the ability to decrease NO production. Additionally, inositol hexaphosphate (IP6), a dietary phytochemical, has demonstrated a reduction in iNOS gene transcription, thus potentially lowering NO levels. Furthermore, NO scavengers like carboxy-2-phenyl-4,4,5,5-tetramethylimidazoline-1-oxyl 3-oxide (c-PTIO) have been effective in targeting NO production from different sources, showing promise in reducing NO-related pathologies. Overall, a combination of natural and synthetic agents, along with NO scavengers, may offer a comprehensive approach to lowering NO levels in the colon and potentially mitigating associated inflammatory conditions.
What are the current protocols used for deriving cells from stem cells for modeling diabetes?
4 answers
Current protocols for deriving cells from stem cells to model diabetes involve generating functional β-cells from human pluripotent stem cells (hPSCs). These protocols focus on aggregating hPSC-derived pancreatic progenitors into spheroids and differentiating them towards an endocrine cell fate in suspension culture, with factors like FGF7 and Notch inhibition playing crucial roles in promoting endocrine cell formation. Additionally, combining multiple hPSC-derived tissues like pancreatic islet cells, T cells, and thymic epithelium has been proposed to model distinct aspects of Type 1 diabetes, providing insights into disease etiology and pathogenesis. Furthermore, the use of mesenchymal stem cells (MSCs), particularly fetal pancreatic-derived MSCs, in diabetes cell-based therapies is being explored, with xeno-free protocols using human serum instead of fetal bovine serum showing promise for clinical-grade MSC manufacturing. These advancements in stem cell applications offer valuable tools for studying diabetes pathophysiology and developing personalized treatments.
What is the role of acidity in tumor cell metabolism?
4 answers
Acidity plays a crucial role in tumor cell metabolism by influencing gene expression, tumor progression, and resistance to therapies. Tumor acidity, resulting from glycolytic metabolism, promotes proliferation, invasiveness, and aggressiveness. Acidosis induces an epithelial-to-mesenchymal transition phenotype, enhancing invasive ability and resistance to apoptosis, resembling stem-like properties in cancer cells. The tumor microenvironment's acidity and hypoxia lead to cancer invasion and progression, with intracellular and extracellular acidity regulation being vital for cell survival in adverse conditions. Additionally, amino acid metabolism, essential for cancer growth and immune response modulation, is influenced by tumor acidity, impacting tumor immunity and therapeutic efficacy. Overall, acidity in tumor cells significantly affects gene expression, functional properties, and immune responses, highlighting its importance in tumor metabolism and progression.